Skip to Main Content
Book cover for Brain's Diseases of the Nervous System (12 edn) Brain's Diseases of the Nervous System (12 edn)

Contents

Book cover for Brain's Diseases of the Nervous System (12 edn) Brain's Diseases of the Nervous System (12 edn)
Disclaimer
Oxford University Press makes no representation, express or implied, that the drug dosages in this book are correct. Readers must therefore always … More Oxford University Press makes no representation, express or implied, that the drug dosages in this book are correct. Readers must therefore always check the product information and clinical procedures with the most up to date published product information and data sheets provided by the manufacturers and the most recent codes of conduct and safety regulations. The authors and the publishers do not accept responsibility or legal liability for any errors in the text or for the misuse or misapplication of material in this work. Except where otherwise stated, drug dosages and recommendations are for the non-pregnant adult who is not breastfeeding.
23.1

Introduction  637

23.1.1

Classification of motor neurone disorders  637

23.1.2

Differential diagnosis  637

23.2

Amyotrophic lateral sclerosis/motor neurone disease 639

23.2.1

Clinical features  639

23.2.2

Diagnostic criteria  643

23.2.3

Differential diagnosis and investigation  643

23.2.4

Pathology  645

23.2.5

Disease pathogenesis  648

23.2.6

Epidemiology  654

23.2.7

Management  654

23.3

Disorders of the lower motor neurone  659

23.3.1

Spinal muscular atrophy  659

23.3.2

Autosomal dominant proximal spinal muscular atrophy  661

23.3.3

Distal spinal muscular atrophy/hereditary motor neuronopathy  661

23.3.4

Monomelic focal and segmental spinal muscular atrophies  662

23.3.5

Hexosaminidase deficiency  663

23.3.6

Multifocal motor neuropathy with conduction block  663

23.3.7

Post-polio progressive muscular atrophy  664

23.4

Disorders of the upper motor neurone  664

23.4.1

Primary lateral sclerosis  664

23.4.2

Hereditary spastic paraplegia  664

23.5

Disorders of the bulbar motor system  672

23.5.1

Kennedy’s disease  672

23.5.2

Brown–Vialetto–van Laere syndrome  673

23.5.3

Fazio–Londe disease  674

23.6

Toxic disorders of the motor neurone  674

23.6.1

Neurolathyrism and Konzo  674

23.6.2

Western Pacific amyotrophic lateral sclerosis-Parkinsonism-dementia complex  675

23.6.3

Post-irradiation motor disorders  675

23.7

Disorders of motor neurone overactivity  676

23.7.1

Neuromyotonia  676

23.7.2

Stiff person syndrome  676

The motor neurone diseases are a group of disorders in which there is selective loss of function of upper and/or lower motor neurones in the motor cortex, brainstem, and spinal cord resulting in impairment in the nervous system control of voluntary movement. The term ‘motor neurone disease’, often abbreviated to ‘MND’, is used differently in different countries. In the United Kingdom it is used as an umbrella term to cover the related group of neurodegenerative disorders including amyotrophic lateral sclerosis, the commonest variant, as well as progressive muscular atrophy, primary lateral sclerosis, and progressive bulbar palsy. However, in many other countries amyotrophi\c lateral sclerosis, referred to as ALS, has been adopted as the umbrella term for this group of clinical variants of motor system degeneration. There is a tendency now internationally to use the ALS/MND abbreviation to cover this group of conditions. Careful diagnosis within the motor neurone diseases is essential for advising about prognosis, potential genetic implications, and for identifying those with acquired lower motor neurone syndromes who may benefit for the administration of immunomodulatory therapy. Table 23.1 classifies the motor neurone disorders according to whether the upper motor neurone, the lower motor neurone, or both groups of cells are affected.

Table 23.1
Classification of the motor neurone disorders

Combined upper and lower motor neurone disorders

Amyotrophic lateral sclerosis (ALS)

Familial adult onset

Familial juvenile onset

Sporadic

ALS-plus syndromes

ALS with frontotemporal dementia

Western Pacific ALS–Parkinsonism–dementia complex

Upper motor neurone disorders

Primary lateral sclerosis

The hereditary spastic paraplegias

Neurolathyrism

Konzo

Lower motor neurone disorders Hereditary

The spinal muscular atrophies (SMAs)

Proximal autosomal recessive SMA of childhood

(associated with SMN mutations)

Type 1 Werdnig Hoffman disease

Type II Intermediate form

Type III Wohlfart–Kugelberg–Welander disease

Type IV Adult onset

Acute infantile forms of SMA not associated with SMN mutations

SMA with pontocerebellar hypoplasia

SMA and arthrogryphosis +/- bone fractures

X-linked SMA with arthogryphosis

Lethal congenital contracture syndromes types 1 and 2

Autosomal dominant proximal SMA

Distal spinal muscular atrophy /hereditary motor neuronopathy

SMA with respiratory distress (SMARD)

Autosomal recessive distal SMA

Distal SMA with upper limb predominance

Distal SMA with lower limb predominance

Distal SMA with vocal cord paralysis

Congenital non-progressive SMA involving the lower limbs

Scapuloperoneal SMA

Kennedy’s disease (X-linked spinobulbar neuronopathy)

Hexosaminidase deficiency (GM2 gangliosidosis)

Acquired

Monomelic focal and segmental spinal muscular atrophies

Multifocal motor neuropathies

Acute motor axonal neuropathy

Post-polio syndrome

Post-irradiation syndrome

Infective disorders

Acute poliomyelitis

West Nile fever

Other viral infections eg enterovirus 71 and rabies

HIV associated motor neurone disorder

Lyme disease

Tick borne encephalitis

Creutzfeld–Jacob disease (amyotrophic forms)

Disorders of the bulbar motor system

Kennedy’s disease (X-linked bulbospinal neuronopathy)

Brown–Vialetto–van Laere syndrome

Fazio Londe disease

Toxic disorders of the motor neuron

Neurolathyrism

Konzo

Heavy metal toxicity (lead, mercury)

Western Pacific ALS–Parkinsonism–dementia complex

Post-irradiation motor neurone injury

Disorders of motor neurone overactivity

Neuromyotonia

Stiff person syndrome

Miscellaneous motor neurone disorders

Endocrinopathies e.g. hyperthyroidism, hyperparathyroidism, hypoglycaemia

Copper deficiency syndrome

Benign cramp-fasciculation syndrome

Combined upper and lower motor neurone disorders

Amyotrophic lateral sclerosis (ALS)

Familial adult onset

Familial juvenile onset

Sporadic

ALS-plus syndromes

ALS with frontotemporal dementia

Western Pacific ALS–Parkinsonism–dementia complex

Upper motor neurone disorders

Primary lateral sclerosis

The hereditary spastic paraplegias

Neurolathyrism

Konzo

Lower motor neurone disorders Hereditary

The spinal muscular atrophies (SMAs)

Proximal autosomal recessive SMA of childhood

(associated with SMN mutations)

Type 1 Werdnig Hoffman disease

Type II Intermediate form

Type III Wohlfart–Kugelberg–Welander disease

Type IV Adult onset

Acute infantile forms of SMA not associated with SMN mutations

SMA with pontocerebellar hypoplasia

SMA and arthrogryphosis +/- bone fractures

X-linked SMA with arthogryphosis

Lethal congenital contracture syndromes types 1 and 2

Autosomal dominant proximal SMA

Distal spinal muscular atrophy /hereditary motor neuronopathy

SMA with respiratory distress (SMARD)

Autosomal recessive distal SMA

Distal SMA with upper limb predominance

Distal SMA with lower limb predominance

Distal SMA with vocal cord paralysis

Congenital non-progressive SMA involving the lower limbs

Scapuloperoneal SMA

Kennedy’s disease (X-linked spinobulbar neuronopathy)

Hexosaminidase deficiency (GM2 gangliosidosis)

Acquired

Monomelic focal and segmental spinal muscular atrophies

Multifocal motor neuropathies

Acute motor axonal neuropathy

Post-polio syndrome

Post-irradiation syndrome

Infective disorders

Acute poliomyelitis

West Nile fever

Other viral infections eg enterovirus 71 and rabies

HIV associated motor neurone disorder

Lyme disease

Tick borne encephalitis

Creutzfeld–Jacob disease (amyotrophic forms)

Disorders of the bulbar motor system

Kennedy’s disease (X-linked bulbospinal neuronopathy)

Brown–Vialetto–van Laere syndrome

Fazio Londe disease

Toxic disorders of the motor neuron

Neurolathyrism

Konzo

Heavy metal toxicity (lead, mercury)

Western Pacific ALS–Parkinsonism–dementia complex

Post-irradiation motor neurone injury

Disorders of motor neurone overactivity

Neuromyotonia

Stiff person syndrome

Miscellaneous motor neurone disorders

Endocrinopathies e.g. hyperthyroidism, hyperparathyroidism, hypoglycaemia

Copper deficiency syndrome

Benign cramp-fasciculation syndrome

In patients with motor neurone diseases, precise differential diagnosis requires clinical and neurophysiological assessment as to whether the disorder involves the upper motor neurones, the lower motor neurones, or both. Consideration should then be given to factors such as age of onset, rapidity of progression, evidence of an inherited disorder, and the anatomical distribution of the clinical features to determine the likely diagnosis. As a general rule, the sensory system, sphincter control, and cognitive function are usually preserved in motor neurone diseases.

The clinical features of lower motor neurone involvement are muscle wasting, fasciculations, and flaccid weakness. Tendon reflexes are usually preserved until the muscle denervation is severe. In amyotrophic lateral sclerosis, the upper motor neurone component tends to preserve the reflexes and the preservation of reflexes in a limb with severe muscular atrophy may be an important clue to the diagnosis. Motor neurone diseases will usually give rise to significant denervation atrophy of weakened muscles. If muscle bulk is relatively preserved in a weak muscle it should raise the possibility of conduction block rather than denervation, as found in multifocal motor neuropathy with conduction block. Fasciculations are visible flickering movements within the muscle belly which are insufficient to produce movement at the joint and are most commonly observed in the large proximal muscle such as deltoid and quadriceps which have large motor units. Fasciculations which are not visible clinically may be detected by electromyography. Fasciculations can only be regarded as indicative of motor neurone disease if associated with evidence of denervation by clinical or electrophysiological assessment.

In patients with suspected motor neurone disease, nerve conduction studies will rule out sensorimotor polyneuropathy; pure motor demyelinating neuropathies or multifocal motor neuropathy with conduction block. Maximal motor conduction velocity is often reduced in nerves supplying denervated muscles as a consequence of degeneration of large motor axons. However, in amyotrophic lateral sclerosis the motor conduction velocity rarely falls below 80 per cent of the lower limit of normal and F waves or distal motor latencies rarely exceed 1.25 times the upper limit of normal. Results outside these limits should raise the possibility of a primary demyelinating motor neuropathy. Electromyography helps to distinguish denervation from myopathy and may also detect subclinical denervation in limbs which are clinically normal in patients with amyotrophic lateral sclerosis. Muscle biopsy may occasionally be required to rule out myopathy, particularly in patients with atypical features or slowly progressive proximal weakness.

Signs of upper motor neurone involvement in motor neuron diseases include increased muscle tone, clonus, pyramidal distribution weakness, and extensor plantar responses. In many patients with amyotrophic lateral sclerosis the presence of clonus and extensor plantar responses is often obscured by the profound denervation changes in the distal lower limb muscles, thereby obscuring clinical confirmation of upper motor neurone involvement. Changes in threshold and parameters of central motor conduction following transcranial magnetic stimulation of the motor cortex may be seen in patients with amyotrophic lateral sclerosis, but the changes observed are complex and may vary with the stage or anatomy of the disease and are insufficiently sensitive to provide a robust and useful tool for detecting subclinical involvement of the upper motor neurone.

The group of patients with amyotrophic lateral sclerosis who do appear to have a pure lower motor neurone disorder without clear clinical signs of upper motor neurone involvement, present a diagnostic challenge. In this situation it is important to exclude treatable lower motor neurone disorders such as multifocal motor neuropathy with conduction block and a trial of immunomodulatory therapy may be indicated where diagnostic uncertainty persists. The passage of time may resolve the diagnostic uncertainty as patients with the pure lower motor neurone variant of amyotrophic lateral sclerosis, known as progressive muscular atrophy, will typically show a brisk rate of disease progression.

The commonest diagnostic problem is to distinguish amyotrophic lateral sclerosis from other motor neurone disorders carrying a better prognosis, particularly if signs of upper motor neurone involvement are lacking. Multifocal motor neuropathy with conduction block usually develops over many years, tends to present with asymmetrical involvement of the upper limbs, may present with difficulty in extending one or more fingers, is associated with electrophysiological evidence of motor nerve conduction slowing or block, and the patient may have antiganglioside antibodies detectable in blood (Section 21.11.3). Patients with post-polio syndrome tend to deteriorate rather slowly by comparison with amyotrophic lateral sclerosis and present with slight further loss of neuromuscular function some decades after an earlier attack of acute poliomyelitis (Section 23.3.7). Kennedy’s disease should be suspected in patients who appear to have amyotrophic lateral sclerosis with bulbar involvement, but who do not deteriorate as rapidly as expected (Section 23.5.1). In men with pure lower motor neurone disorders the presence of gynaecomastia should always be looked for. Other diagnostic alerts for Kennedy’s disease include the presence of mentalis contractions or fasciculations and clinical or neurophysiological evidence of sensory nerve involvement.

Muscle fasciculations are common in the normal population and are frequently observed in the calf muscles after exercise. Patients with benign fasciculations who come to neurological attention are frequently individuals with medical knowledge who fear the development of motor neurone disease, or those with a family history of amyotrophic lateral sclerosis. Benign fasciculations are most commonly observed in the calf muscles, are not associated with any other abnormal clinical signs and electromyography shows no evidence of denervation.

Historical descriptions of amyotrophic lateral sclerosis, or ALS, were reported as early as 1824 by Charles Bell and others (Goldblatt 1968; Tyler and Sheffner 1991). However, Charcot was the first to describe the condition in detail, based on careful clinico-pathological correlation and he coined the term amyotrophic lateral sclerosis (Charcot and Joffroy 1869). Motor neurone disease, or MND, as used by Brain in the first edition of this textbook (Brain 1933), is the name used for the disease most commonly in the United Kingdom and represents an umbrella term encompassing amyotrophic lateral sclerosis and three other disorders that are considered its clinical variants: primary lateral sclerosis, progressive muscular atrophy, and progressive bulbar palsy. In the USA, amyotrophic lateral sclerosis is sometimes known as Lou Gehrig’s disease after the famous Yankee baseball player who developed the disease at the peak of his sporting career and died in 1941. Amyotrophic lateral sclerosis is sometimes referred to as ALS/MND.

Amyotrophic lateral sclerosis is a neurodegenerative disorder that causes progressive injury and cell death of lower motor neurones within the brainstem and spinal cord and upper motor neurones in the motor cortex. The disease has an incidence of about 2 per 100 000 and a prevalence of 6–8 /100 000 (Chancellor and Warlow 1992; Traynor et al. 1999). There are approximately 5000 individuals with amyotrophic lateral sclerosis at any one time in the UK. The incidence is fairly uniform throughout the world, with the exception of a few high incidence foci, for example, on the Kii peninsula of Japan and the Western Pacific island of Guam. The disease predominantly affects middle-aged and elderly individuals, with a mean age of onset of 55 years, though younger individuals can also be affected. For reasons that are not understood, men are affected more commonly than women, with a male/female ratio of approximately 1.6/1. Motor neurone disease is sporadic in 90 per cent of cases, but in 5–10 per cent of cases the disease is familial, usually with an autosomal dominant mode of inheritance.

Amyotrophic lateral sclerosis is characterized and defined by the presence of clinical features reflecting degeneration of upper motor neurones of the cerebral cortex, and lower motor neurones of the brainstem and spinal cord. Lower motor neurone degeneration causes weakness, atrophy, and fasciculation of the limb and bulbar musculature. Muscle cramps are a common symptom, and patients may be aware of fasciculation as twitching or flickering movements of their muscles. Clinical signs resulting from degeneration of the upper motor neurones include the incongruous presence of active or brisk tendon reflexes in a wasted limb, increased muscle tone, and sometimes the presence of Hoffman’s or Babinski’s signs. Upper motor neurone dysfunction within the bulbar territory causes pseudobulbar palsy, where the snout and jaw reflexes may be exaggerated, and increased tone within the bulbar muscles may cause slowing of repetitive movements of the tongue as well as strained effortful speech. Emotional lability, producing difficulty in controlling episodes of laughing or crying, often accompanies upper motor neurone signs in the bulbar region.

In amyotrophic lateral sclerosis the disease often starts focally and asymmetrically in the upper limb, lower limb, or bulbar territories. This is followed by progressive spread of injury to contiguous groups of motor neurones, so that the patient’s clinical features often show an anatomically logical progression. By the end of the disease course most patients will have features of upper and lower motor neurone dysfunction affecting all four limbs and the bulbar musculature.

The first clinical problem is present in the upper or lower limbs in approximately 75 per cent of patients. At first asymmetry or unilaterality of symptoms and signs is common and the first noticeable problem for the patient commonly involves the distal limb muscles. Affected individuals may notice weakness, wasting or clumsiness of one hand, or simply difficulty with everyday actions such as turning a key or lifting a heavy object. In the lower limbs, foot drop or a tendency to trip over the toes of one foot is a common presenting symptom. The gait may become slowed or clumsy due to weakness and spasticity of one or both lower limbs. The patient may be aware of limb muscle cramps which can sometimes precede other clinical features of amyotrophic lateral sclerosis by months or years. In addition, muscle fasciculation may be noticed by the patient, particularly arising from the large proximal limb muscles such as biceps, triceps, pectoralis major, and quadriceps. As the condition progresses, patients often develop a characteristic pattern of limb muscle weakness. In the upper limbs, the intrinsic hand muscles, particularly the thenar group, tend to be affected severely and early, whereas other muscle groups such as triceps and the finger flexors are relatively spared until late in the disease course. In the lower limbs, the pattern of weakness is often in a pyramidal distribution, with flexors weaker than extensors, early weakness of hip flexion and ankle dorsiflexion and often more severe weakness of the distal muscles. Examination of the patient will frequently reveal a combination of upper and lower motor neurone features with proximal fasciculation, muscle wasting, particularly distally (Fig. 23.1), the characteristic pattern of weakness described above, but in addition tone may be mildly increased, the reflexes are brisk and extensor plantar responses may be present.

 Wasting of the intrinsic hand muscles in a patient with amyotrophic lateral sclerosis.
Fig. 23.1

Wasting of the intrinsic hand muscles in a patient with amyotrophic lateral sclerosis.

Bulbar symptoms are the presenting clinical problem in approximately 25 per cent of patients with amyotrophic lateral sclerosis and this presentation is particularly common in elderly women. The onset of amyotrophic lateral sclerosis in the bulbar regions has a less favourable prognosis than limb-onset disease (Chancellor et al. 1993; del Aguila et al. 2003). The first problem is usually slurring of speech which initially may only be apparent when the individual is tired at the end of the day, after prolonged use of the voice, or after alcohol. The dysarthria may rarely be accompanied by dysphonia. The patient may notice particular difficulty with specific sounds such as ‘s’ and may notice other features of bulbar muscle weakness such as difficulty in pursing the lips or whistling. The onset of dysarthria is often initially attributed to a ‘minor stroke’, but it soon becomes apparent that the speech difficulty is a progressive problem. Patients with amyotrophic lateral sclerosis often have a mixed spastic/flaccid dysarthria. The speech develops a tight, strangled quality due to the upper motor neurone component, but superimposed on this, the flaccid lower motor neurone weakness of the palate and nasopharynx give the speech a nasal quality. Examination of the patient will often reveal a combination of upper and lower motor neurone bulbar signs with weakness of the facial

muscles, a spastic, weak, wasted, and fasciculating tongue and a brisk jaw jerk. As the disease progresses the patient may develop weakness of the palate and muscles of mastication. Occasionally forcible jaw clenching is a troublesome symptom causing trauma to the mouth. Dysphagia becomes a problem later, often several weeks or months after the onset of the speech problem. Initially dysphagia tends to be more pronounced for liquids compared to solids. Gradually certain food items such as lettuce, foods with a crumbly texture, or items with a sharp taste become difficult to swallow and may cause choking episodes. Mealtimes may become prolonged and arduous, with frequent episodes of coughing. When dysphagia reaches a significant level, the patient will have a problem with excess saliva pooling in the mouth. If accompanied by lower facial weakness, the patient will suffer from drooling which causes considerable social embarrassment. As the bulbar dysfunction becomes increasingly severe, the patient may develop complete anarthria. Severe dysphagia causes the patient to be at risk of aspiration and chest infection.

Weight loss. This is a common feature of amyotrophic lateral sclerosis. Multiple factors may contribute to this, with loss of muscle bulk due to amyotrophy, difficulty maintaining nutrition in the face of dysphagia, and loss of appetite resulting from reactive anxiety and depression or from immobility. Severe weight loss and nutritional deficiencies may themselves exacerbate muscle weakness.

Neck muscle weakness. This is common later in the course of disease, causing difficulty holding the head upright, the ‘dropped head syndrome’. This problem commonly causes pain and increases difficulties with swallowing and communication.

Respiratory muscle weakness. Occasionally, onset of the disease in motor neurones innervating the respiratory muscles causes breathlessness as a presenting symptom (Nightingale et al. 1982; De Carvalho et al. 1996). More commonly, respiratory failure develops insidiously during the course of the disease, causing dyspnoea and orthopnoea. Significant diaphragmatic weakness may be apparent from paradoxical movement of the abdominal wall during inspiration and from a marked decline in the forced vital capacity when measured in the supine compared to the upright position. Symptoms of nocturnal carbon dioxide retention may develop, including morning headaches, anorexia, and daytime somnolence.

Extramotor features. Amyotrophic lateral sclerosis is traditionally considered to be a pure motor disorder, which spares sensation, cognition, and autonomic function. However, there is increasing evidence that, despite the relative vulnerability of motor neurones to the degenerative process, other types of neurones are also affected. Thus, amyotrophic lateral sclerosis is now regarded as a multisystem neurodegenerative disorder in which the earliest and most severe degeneration tends to involve motor neurones. In most patients, the evolution of motor dysfunction is lethal before the development of overt signs of central nervous system pathology in other regions, although occasional patients may spontaneously develop a severe multisystem degeneration (Machida et al. 1999). Patients whose survival is prolonged by ventilatory support may develop widespread features of extramotor system involvement.

Cognitive impairment. Frontotemporal dementia (Section 34.6.5) encompasses a range of clinical features that result from atrophy of the frontal and anterior temporal regions of the brain. In the frontal variant of frontotemporal dementia, there is prominent personality change and conduct disorder, and affected individuals often become disinhibited or apathetic with emotional blunting, and loss of insight. Behaviour tends to become ritualized or stereotypical, and eating behaviour often becomes abnormal. Visuo-spatial function and memory are relatively preserved. Two syndromes of progressive language dysfunction are seen with frontotemporal dementia. Primary progressive aphasia, which may progress to mutism, is characterized by effortful speech production with phonological and grammatical errors, and word retrieval difficulties. Patients may also develop semantic dementia, with impairment of naming and word comprehension but preservation of fluent, grammatical speech output (Neary et al. 1998; McKhann et al. 2001; Hodges et al. 2004). There is overlap between these clinical syndromes, as patients with either syndrome may later develop deterioration in personality and behaviour (Bak et al. 2001), while language abnormalities may be found in patients with frontotemporal dementia (Neary et al. 1990; Strong et al. 1996).

Approximately 5 per cent of patients with motor neurone disease will develop overt features of frontotemporal dementia (Hudson 1981). Cognitive dysfunction may precede, follow, or coincide with the features of motor dysfunction. The most common symptoms seen in frontotemporal dementia in association with motor neurone disease are progressive deterioration in personality and behaviour (Ringholz et al. 2005), but primary progressive aphasia is also described.

Motor neurone disease patients without overt dementia may show more subtle features of frontal lobe dysfunction. This problem may be under-recognized in the normal clinic setting, partly because of the difficulty in assessing cognitive function in patients with bulbar dysfunction and severe motor deficits. Detailed neuropsychological assessment demonstrates that up to 50 per cent of patients with sporadic motor neurone disease develop features of frontal lobe dysfunction (Ringholz et al. 2005), There is evidence indicating that cognitive impairment may be more common in patients with primary lateral sclerosis or predominant upper motor neurone signs (Caselli et al. 1995), and in those with prominent bulbar involvement (Schreiber et al. 2005).

Parkinsonism. This is described in cases of sporadic motor neurone disease, and occurs more frequently in cases with accompanying dementia (Qureshi et al. 1996). The prevalence of Parkinsonism in motor neurone disease may be underestimated, as extrapyramidal features can be masked in patients with muscle weakness, wasting, and spasticity. Subclinical defects in dopaminergic transmission have been demonstrated by positron emission tomography scanning in patients with sporadic motor neurone disease, without clinical evidence of an extrapyramidal disorder (Takahashi et al. 1993).

Disorders of eye movement. Supranuclear ophthalmoplegia, including limitation of ocular movement, slow ocular movement, and spasmodic gaze fixation, have been described in motor neurone disease. Although the majority of these patients develop these features after a disease course extended by invasive ventilation, they can occasionally be seen in ‘natural disease’ (Hayashi et al. 1989; Mizutani et al. 1990). Difficulty initiating eyelid opening, with preserved reflex eyelid movements, often referred to as ‘eyelid apraxia’, is also seen in patients with motor neurone disease, and may be associated with supranuclear vertical gaze impairment (Abe et al. 1995).

Sensory impairment. Patients with amyotrophic lateral sclerosis not infrequently complain of non-specific sensory phenomena, such as tingling in the fingers. Sensory examination is usually normal, but nerve conduction studies and somatosensory-evoked potentials reveal the presence of abnormalities in peripheral and central sensory pathways in up to 60 per cent of patients (Bosch et al. 1985; Shefner et al. 1991). There are isolated case reports of more profound sensory involvement in association with amyotrophic lateral sclerosis (Wakabayashi et al. 1998).

Selective sparing of specific motor neurone groups. The eye movements tend to be spared in motor neurone disease. Even in advanced disease, when patients would otherwise be ‘locked in’, often limited communication can be retained by movements of the eyes. Similarly, the strength of the pelvic floor muscles is relatively preserved, so that patients with motor neurone disease usually remain continent throughout the course of the disease. These selectively spared muscle groups reflect the fact that motor neurones in the oculomotor nuclei of the brainstem and Onuf’s nucleus in the sacral spinal cord are less vulnerable to the pathological process in motor neurone disease compared to those innervating limb and bulbar musculature.

There are several clinical variants that describe the predominant clinical features of the patient at the time of presentation. As the disease progresses, however, the majority of patients will develop features of upper and lower motor neurone degeneration affecting the limbs and bulbar musculature, the most common variant of amyotrophic lateral sclerosis.

Progressive muscular atrophy. Approximately 5–10 per cent of patients with motor neurone disease will present with clinical features reflecting only degeneration of lower motor neurone groups in the spinal cord, in the absence of any evidence of upper motor neurone pathology. Some of these patients will later develop brisk reflexes or extensor plantar responses. Clinical examination may not reliably detect upper motor neurone involvement late in the disease, when there is severe global weakness and amyotrophy. It is noteworthy that in those patients who develop no clinical upper motor neurone signs during life, approximately 50 per cent will have pathological evidence of corticospinal tract pathology at autopsy (Ince et al. 2003). This suggests that progressive muscular atrophy is part of the same pathological spectrum of disease as amyotrophic lateral sclerosis.

Primary lateral sclerosis. Primary lateral sclerosis, first described by Erb (1875), is a degenerative disorder of the upper motor neurone pathways causing spasticity of the limb and bulbar muscles. It is usually a sporadic disorder of insidious onset, and commonly starts in the fifth decade or later as a spastic paraparesis. Bulbar or upper limb onset has also been described. The course is gradually progressive, and although patients ultimately develop a severe spastic spinobulbar paresis, survival is usually prolonged compared to patients with classical amyotrophic lateral sclerosis (Pringle et al. 1992). Many patients with primary lateral sclerosis will survive more than 10 to 15 years after symptom onset.

Not all case series of primary lateral sclerosis have adequately excluded alternative diagnoses, particularly prior to the availability of modern imaging techniques. Pringle and colleagues described a series of 8 patients with progressive symmetrical spinobulbar spasticity in whom alternative diagnoses were carefully excluded, and proposed diagnostic criteria for primary lateral sclerosis (Pringle et al. 1992). Although the cardinal feature of primary lateral sclerosis is upper motor neurone involvement, in most case series described, there is usually evidence clinically or electrophysiologically of some lower motor neurone dysfunction, which may only develop after several years (Kuipers-Upmeijer et al. 2001; Le Forestier et al. 2001). The evolution of primary lateral sclerosis into amyotrophic lateral sclerosis has been described, with the onset of generalized amyotrophy developing after many years of a slowly progressive degeneration of the upper motor neurone system, (Bruyn et al. 1995), and primary lateral sclerosis may occur as a phenotypic manifestation of familial amyotrophic lateral sclerosis. These findings strongly indicate that primary lateral sclerosis is part of the spectrum of amyotrophic lateral sclerosis, with predominant but not exclusive degeneration of upper motor neurones.

Progressive bulbar palsy. Bulbar onset motor neurone disease occurs in approximately 25 percent of patients and is most common in women of middle to elderly age. Progressive bulbar palsy usually progresses to involve the limbs, although clinical and electrophysiological abnormalities affecting the limbs may not be found at the time of presentation.

Segmental variants. Several variants of amyotrophic lateral sclerosis have been described, in which the disease follows a more segmental pattern than is typical in classical disease. The flail arm syndrome, progressive amyotrophic diplegia, may occur in up to 10 per cent of patients with motor neurone disease (Fig. 23.2) (Gamez et al. 1999; Katz et al. 1999). It is much more common in men than

 The flail arm syndrome variant of amyotrophic lateral sclerosis.
Fig. 23.2

The flail arm syndrome variant of amyotrophic lateral sclerosis.

in women, and has a longer median survival than classical amyotrophic lateral sclerosis (Hu et al. 1998). It is characterized by profound symmetrical weakness and wasting of the upper limbs with hyporeflexia. Although signs of pyramidal tract involvement may be seen in the lower limbs, there is little or no functional impairment of the bulbar muscles or legs at presentation, and these regions of the motor system are usually involved only late in the disease course. A similar focal presentation in the lower limbs with progressive paraparesis is recognized. Other forms of segmental motor neurone degeneration are described in Section 23.3.4.

Madras form. A specific subtype of early onset sporadic amyotrophic lateral sclerosis was identified in Southern India in 1970 (Meenakshisundaram et al. 1970; Saha et al. 1997; Gourie-Devi and Nalini 2003b). The phenotype is that of young onset amyotrophic lateral sclerosis with bulbar and limb involvement. Lower motor neurone features tend to predominate, but upper motor neurone signs are present and include brisk tendon reflexes and extensor plantar responses. The onset is usually in the second or third decades and the disorder is more common in males. Typical early features comprise distal upper limb weakness with wasting, dysarthria, dysphagia, and progressive sensorineural hearing loss. Progressive deafness is found in more than 50 per cent of affected individuals and is an important feature distinguishing Madras amyotrophic lateral sclerosis from other forms with early sporadic onset. Spasticity and distal leg weakness frequently develop and the disease course is one of progressive disability but with long-term survival. Electromyography studies show chronic partial denervation. Pathological findings from one case have been described (Shankar et al. 2000). There was depletion of lower motor neurones from the spinal cord and brainstem; neuronal loss and gliosis in the cochlear nucleus; demyelination and axonal loss in the cochlear nerve, and degeneration of the corticospinal tracts with accompanying gliosis. There was no description of whether ubiquitinated inclusion bodies were present in surviving motor neurones.

Symptoms and signs are often focal at the time the patient with amyotrophic lateral sclerosis first presents to medical attention, though neurophysiological evidence of motor neurone injury may be more widespread. In the majority of cases there is an inexorable progression of the pathological process, often seemingly to contiguous groups of motor neurones. At the end stage of the disease, the patient will usually have significant motor dysfunction affecting all four limbs and the bulbar muscles as well as compromise of respiratory function. The patient may become entirely dependent upon nursing care, bulbar dysfunction may progress to anarthria, and enteral feeding may be required due to severe dysphagia.

Certain clinical features are associated with a worse prognosis for the clinical course of the disease including: older age at onset of symptoms; early compromise of respiratory function; bulbar onset of symptoms and more rapid presentation of the patient to medical attention (Chancellor et al. 1993; del Aguila et al. 2003; Millul et al. 2005). The mean survival from symptom onset is approximately 3 years, although the rate of progression varies between individuals. Rapid variants of disease may progress to death within a few months, but approximately 4 per cent of patients will survive for more than 10 years (Turner et al. 2003). The usual cause of death is progressive respiratory failure, which may be accompanied by bronchopneumonia.

The El Escorial Diagnostic criteria for amyotrophic lateral sclerosis were agreed as an international consensus to facilitate therapeutic trials and multinational research collaborations (Brooks 1994). These criteria were subsequently revised and the key features of the revised criteria are shown in Table 23.2 (Brooks et al 2000). In essence there must be a combination of upper and lower motor neurone signs, evidence of progression over at least 6 months, and other conditions that may mimic amyotrophic lateral sclerosis must be excluded by appropriate investigations. These criteria are stricter than the burden of proof for the diagnosis of amyotrophic lateral sclerosis usually applied in clinical practice and indeed some individuals die from the effects of amyotrophic lateral sclerosis without ever reaching the classification criteria required for definite diagnosis (Traynor et al. 2000b). Clinicians should be cautious about applying these criteria in clinical practice as patients may be confused and less able to come to terms with their illness by being told that they have probable or possible amyotrophic lateral sclerosis rather than a more definite diagnosis. Nevertheless, the criteria provide a structured approach to the evaluation of patients with amyotrophic lateral sclerosis and facilitate the inclusion of uniform populations of patients in clinical research studies.

Table 23.2
Summary of revised El Escorial research diagnostic criteria for amyotrophic lateral sclerosis (ALS)

The diagnosis of amyotrophic lateral sclerosis requires:

(A:1) Evidence of LMN degeneration by clinical, electrophysiological, or neuropathological examination;

(A:2) Evidence of UMN degeneration by clinical examination, and

(A:3) Progressive spread of symptoms or signs within a region or to other regions, as determined by history or examination,

Together with

(B) The absence of:

(B:1) Electrophysiological and pathological evidence of other disease that might explain the signs of LMN and/or UMN degeneration, and

(B:2) Neuroimaging evidence of other disease processes that might explain the observed clinical and electrophysiological signs

Within these principles, one can diagnose:

Definite ALS

UMN signs and LMN signs in three regions

Probable ALS

UMN signs and LMN signs in two regions with at least some UMN signs rostral to LMN signs

Probable ALS—Laboratory supported

UMN signs in one or more regions and LMN signs defined by EMG in at least two regions

Possible ALS

UMN signs and LMN signs in one region (together), or

♦ UMN signs in two or more regions

♦ UMN and LMN signs in two regions with no UMN signs rostral to LMN signs

The diagnosis of amyotrophic lateral sclerosis requires:

(A:1) Evidence of LMN degeneration by clinical, electrophysiological, or neuropathological examination;

(A:2) Evidence of UMN degeneration by clinical examination, and

(A:3) Progressive spread of symptoms or signs within a region or to other regions, as determined by history or examination,

Together with

(B) The absence of:

(B:1) Electrophysiological and pathological evidence of other disease that might explain the signs of LMN and/or UMN degeneration, and

(B:2) Neuroimaging evidence of other disease processes that might explain the observed clinical and electrophysiological signs

Within these principles, one can diagnose:

Definite ALS

UMN signs and LMN signs in three regions

Probable ALS

UMN signs and LMN signs in two regions with at least some UMN signs rostral to LMN signs

Probable ALS—Laboratory supported

UMN signs in one or more regions and LMN signs defined by EMG in at least two regions

Possible ALS

UMN signs and LMN signs in one region (together), or

♦ UMN signs in two or more regions

♦ UMN and LMN signs in two regions with no UMN signs rostral to LMN signs

UMN: (upper motor neurone) signs: clonus, Babinski sign, absent abdominal skin reflexes, hypertonia, loss of dexterity.

LMN: (lower motor neurone) signs: atrophy, weakness. If only fasciculation: search with EMG (electromyography) for active denervation.

Regions reflect segmental neuronal pools: bulbar, cervical, thoracic, and lumbosacral.

The main conditions to be considered in the differential diagnosis of amyotrophic lateral sclerosis are listed in Table 23.3. Diagnostic errors are not uncommon in amyotrophic lateral sclerosis and an important aspect of follow-up care is to review the diagnosis in patients whose symptoms or disease course have atypical features. Of crucial importance is not to miss the diagnosis of potentially treatable disorders or those with a more benign prognosis. In a population-based survey from Scotland (Davenport et al. 1996) amyotrophic lateral sclerosis was misdiagnosed in 8 per cent of 552 patients included on the register. In an Irish population-based study which reported 437 patients’ referrals diagnosed initially as amyotrophic lateral sclerosis, an alternative diagnosis was found in 7 per cent (Traynor et al. 2000a). Most of the misdiagnosed cases had lower motor neurone syndromes: 22 per cent had multifocal motor neuropathy with conduction block (Sections 23.3.6 and 21.11.3) and 13 per cent had Kennedy’s disease (Section 23.5.1). Other forms of motor neuropathy should also be considered including acute motor axonal neuropathy (Section 21.10.2), a variant of Guillain–Barré syndrome (Griffin et al. 1996; Léger and Salachas 2001) and acute porphyric neuropathy (Section 21.8.6). Myasthenia gravis (Section 24.10.1), particularly with prominent bulbar involvement, may occasionally mimic features of amyotrophic lateral sclerosis, though upper motor neurone clinical signs will be absent.

Table 23.3
Differential diagnosis of amyotrophic lateral sclerosis/motor neurone disease

Other acquired motor neuropathies including multifocal motor neuropathy with conduction block, acute motor axonal neuropathy, and acute porphyric neuropathy.

Kennedy’s disease

Myasthenia gravis

Benign fasciculation syndromes

Peripheral nerve hyperexcitability with myokymia

Focal mechanical lesions of the spinal cord or brainstem e.g. multilevel spondylotic radiculomyelopathy, syringomyelia, tumours of base of skull, spinal cord, cauda equina/conus medullaris

Hereditary spastic paraplegia

Adult onset spinal muscular atrophy and hereditary motor neuropathies

Myopathies including inclusion body myositis and polymyositis

Multisystem neurodegenerative disorders e.g. frontotemporal dementia; multisystem atrophy; progressive supranuclear palsy; corticobasal degeneration; spinocerebellar atrophies (SCA 6 and SCA3)

Infective disorders eg HTLVI; HIV; Lyme disease

Post-polio syndrome

Paraneoplastic syndromes

Metabolic disorders e.g. hyperthyroidism and hyperparathyroidism

Other acquired motor neuropathies including multifocal motor neuropathy with conduction block, acute motor axonal neuropathy, and acute porphyric neuropathy.

Kennedy’s disease

Myasthenia gravis

Benign fasciculation syndromes

Peripheral nerve hyperexcitability with myokymia

Focal mechanical lesions of the spinal cord or brainstem e.g. multilevel spondylotic radiculomyelopathy, syringomyelia, tumours of base of skull, spinal cord, cauda equina/conus medullaris

Hereditary spastic paraplegia

Adult onset spinal muscular atrophy and hereditary motor neuropathies

Myopathies including inclusion body myositis and polymyositis

Multisystem neurodegenerative disorders e.g. frontotemporal dementia; multisystem atrophy; progressive supranuclear palsy; corticobasal degeneration; spinocerebellar atrophies (SCA 6 and SCA3)

Infective disorders eg HTLVI; HIV; Lyme disease

Post-polio syndrome

Paraneoplastic syndromes

Metabolic disorders e.g. hyperthyroidism and hyperparathyroidism

Benign fasciculation. This is relatively common, affecting approximately 1 per cent of the population, and often causes great anxiety, particularly in individuals with medical knowledge. The syndrome comprises chronic muscle fasciculation, most commonly in the calf muscles, which may be accompanied by cramps and is often worsened by exercise. Fasciculation may be detected by electromyography, but is not accompanied by evidence of progressive denervation (Blexrud et al. 1993). One study describing 121 patients with a diagnosis of benign fasciculation, 33 per cent of them health workers, seen at the Mayo clinic, reported no cases developing amyotrophic lateral sclerosis during a prolonged period of follow-up (Blexrud et al. 1993). However, individuals presenting with fasciculations and cramps may very rarely go on to develop amyotrophic lateral sclerosis (de Carvalho and Swash 2004). Muscle cramps and twitching in the presence or absence of weakness may occur in peripheral nerve hyper-excitability with myokymia (Hart et al. 2002; Gutmann and Gutmann 2004; Lagueny 2005). Myokymia usually consists of rippling continuous muscle contractions, rather than the discrete random twitching typically seen with fasciculation.

Focal structural lesions. These lesions of the brainstem or spinal cord may sometimes be confused with amyotrophic lateral sclerosis, particularly multilevel spondylotic radiculomyelopathy which may produce upper motor neurone signs accompanied by lower motor neurone signs in the upper and lower limbs. Other structural lesions which may occasionally be mistaken for amyotrophic lateral sclerosis include mass lesions at the skull base; slowly growing tumours of the cervical spinal cord, cauda equina or conus medullaris, and syringomyelia. Many of these patients will have pain and sensory disturbance as well as motor dysfunction. Other more peripheral lesions such as infiltration or compression of the lower roots of the brachial plexus by a Pancoast tumour (Section 22.4.2) or a cervical rib (Section 22.5.3) will also usually be accompanied by pain and sensory features. Occasionally patients with infiltrative lesions of the base of tongue may mimic the features of bulbar onset amyotrophic lateral sclerosis and the accompanying pain is an important diagnostic clue. In the presence of these structural lesions electrophysiological and imaging studies will usually lead to the correct diagnosis.

Hereditary spastic paraplegia variants (Section 23.4.2). These may have lower motor neurone features as well as the typical slowly progressive upper motor neurone disorder primarily affecting the lower limbs. There is clinical overlap between hereditary spastic paraplegia and the primary lateral sclerosis variant of motor neurone disease (Brugman et al. 2005; Strong and Gordon 2005). Patients with the Troyer syndrome, SPG 20, and Silver syndrome, SPG 17, typically have both upper and lower motor neurone features, but a much more slowly progressive clinical course than typical amyotrophic lateral sclerosis. Adult onset forms of spinal muscular atrophy and hereditary motor neuronopathy can pose diagnostic difficulties. Individuals with apparently sporadic lower motor neurone syndromes who survive more than 4 years from the onset of symptoms may turn out to have relatively benign forms of late onset spinal muscular atrophy (van den Berg-Vos et al. 2003a,b) (Section 23.3). Clues to the diagnosis of hereditary motor neuronopathy or spinal muscular atrophy include slow progression, family history, pes cavus, and other features such as vocal cord involvement.

Inclusion body myositis (Section 24.7.3) may mimic the progressive muscular atrophy variant. This is one of the most common types of myopathy presenting in those over 50 years of age (Griggs et al. 1995; Dalakas 2006). In one series of 70 patients with inclusion body myositis, 13 per cent had originally received a diagnosis of amyotrophic lateral sclerosis (Dabby et al. 2001). Important clues to the diagnosis of inclusion body myositis include early and prominent weakness of finger flexors and quadriceps muscles, which tend only to become weak late in the course of amyotrophic lateral sclerosis. Dysphagia may occur. Re-examination of patients both clinically and by electrophysiology where there is diagnostic uncertainty is very important and muscle biopsy may sometimes be indicated. Polymyositis may sometimes be misdiagnosed as amyotrophic lateral sclerosis. For example one reported case of biopsy-proven polymyositis was reported to show clinical features including dysphagia, possible upper motor neurone signs, normal creatine kinase level, and fasciculations on electromyography, causing understandable diagnostic uncertainty (Ryan et al. 2003).

Multisystem degenerative disorders. Disorders in which features of upper and lower motor neurone damage may occur include the frontotemporal dementia syndromes; subtypes of spinocerebellar atrophy including SCA6 and SCA3 (Section 39.8) (Ohara et al. 2002; Seilhean et al. 2004), multiple system atrophy (Section 40.3.8), progressive supranuclear palsy (Section 40.3.9), and corticobasal degeneration (Section 40.3.10) (Neary et al. 1990; Strong et al. 2003; Kertesz et al. 2005; Mott et al. 2005). These multisystem disorders all have prominent features of involvement of the central nervous system outside the upper and lower motor neurones including involvement of basal ganglia, the cerebellum, and cortical structures, so that diagnostic confusion with amyotrophic lateral sclerosisis is uncommon.

Infective. Infective and post-infective disorders occasionally need to be considered in the differential diagnosis of amyotrophic lateral sclerosis. HTLV1 associated myelopathy (Sections 28.5.8; 42.4.1) typically produces a slowly progressive spastic paraparesis with early involvement of the bladder. It has been reported to occasionally be associated with amyotrophic lateral sclerosis-like features, with wasting and fasciculation of the tongue and limbs and electrophysiological evidence of widespread denervation (Matsuzaki et al. 2000; Silva et al. 2005). Patients have been reported with an amyotrophic lateral sclerosis-like disorder in the presence of HIV infection and some patients have apparently improved neurologically with highly active anti-retroviral treatment, ‘HARRT’ (Moulignier et al. 2001; Calza et al. 2004). Often the motor disorder occurring in the context of HIV is not entirely typical of amyotrophic lateral sclerosis, with a younger age of onset and a more rapidly progressive disease. The post-polio progressive muscular atrophy syndrome (Section 23.3.7) may sometimes be mistaken for amyotrophic lateral sclerosis, but its clinical course is much more slowly progressive. It may affect up to one-third of individuals who suffered acute paralytic poliomyelitis and is usually regarded as a syndrome of motor decompensation related to ageing (Ramlow et al. 1992; Ragonese et al. 2005). Lyme disease (Section 42.5.2) has been described as causing a progressive motor neurone disorder which may improve following the administration of antibiotic therapy (Hemmer et al. 1997). Lyme disease is not established as a cause of amyotrophic lateral sclerosis, but it is reasonable to consider the condition as a potential mimicking syndrome in individuals with exposure to tick bites, or when the CSF contains oligoclonal immunoglobulin bands. It is noteworthy that antibodies to Borrelia burgdorferi are commonly encountered in people living in endemic areas (Halperin et al. 1990). Shoulder girdle amyotrophy is a feature of infection by the tick borne encephalitis virus (Logina et al. 2006).

Malignancy. Most association studies have failed to demonstrate a convincing link between amyotrophic lateral sclerosis and malignant disease (Section 38.4.4) (Jokelainen 1976; Evans et al. 1990; Rosenfeld and Posner 1991; Freedman et al. 2005). However, some reports have suggested a possible association with several types of haematological malignancy and melanoma (Younger et al. 1990; Rowland et al. 1995; Gordon et al. 1997; Freedman et al. 2005). An amyotrophic lateral sclerosis-like syndrome has been reported in association with anti-Hu antibodies, and breast cancer may be linked to a primary lateral sclerosis like disorder without anti-Hu antibodies (Forsyth et al. 1997). Predominantly motor neuropathies or neuronopathies can be associated with anti-Hu or anti-Yo antibodies (Khwaja et al. 1998; Graus et al. 2001). Overall, a causative association between amyotrophic lateral sclerosis and cancer is not generally regarded as proven, although there are occasional reports of remission of motor dysfunction following therapy for malignant disease.

Metabolic disorders. These disorders occasionally mimic the features of amyotrophic lateral sclerosis. Hyperthyroidism may present with muscle weakness, wasting, and fasciculation (Rosati et al. 1980; Chotmongkol 1999). Some patients have superimposed upper motor neurone signs, a combination resulting in a clinical picture similar to that of amyotrophic lateral sclerosis (Fisher et al. 1985; Shaw et al. 1988). Treatment of the hyperthyroid state usually results in complete or near complete recovery of the upper motor neurone signs. Hyperparathyroidism may present with weakness and brisk reflexes and a causal relationship between hyperparathyroidism has been suggested (Patten and Pages 1984), but not established with certainty (Jackson et al. 1998).

The diagnosis of amyotrophic lateral sclerosis is essentially clinical and there is no specific diagnostic test. Neurophysiological evaluation and MRI of the brain and spine are the most useful investigations. Sensory nerve conduction is usually normal and motor nerve conduction velocity is also normal unless there has been severe depletion of large diameter motor axons. It is important that conduction block is carefully excluded and this may require evaluation of proximal nerve segments. Electromyography is valuable in demonstrating neurogenic changes that cannot be explained by a single nerve, root, or plexus lesion (Mills 2003). Assessment of the thoracic paraspinal muscles can be particularly valuable in differentiating amyotrophic lateral sclerosis from multilevel spondylotic radiculomyelopathy (Kuncl et al. 1988). Imaging is useful to exlude the presence of structural disorders. A variety of blood tests may be helpful in distinguishing the amyotrophic lateral sclerosis mimic syndromes outlined above. Muscle biopsy is only indicated in rare or atypical cases where diagnostic uncertainty persists in the light of the initial investigation results.

Motor neurone disease has been considered as traditionally a pure motor disorder, but the selectivity of the disease process for the motor system is now recognized to be relative rather than absolute. Careful clinical and pathological studies have revealed involvement in extra-motor parts of the central nervous system such as changes in other long tracts, including sensory and spinocerebellar pathways, and in neuronal groups such as substantia nigra neurones and dentate granule cells in the hippocampus. The description of ubiquitinated intraneuronal inclusions in 1988 highlighted a common molecular pathology in motor neurone disease (Leigh et al. 1988; Lowe et al. 1989) and has provided evidence of widespread involvement of extramotor regions of the central nervous system. Furthermore, the ubiquitinated inclusion is described in a number of related disorders, including primary lateral sclerosis, progressive muscular atrophy, and frontotemporal dementia, supporting the hypothesis that these conditions represent a clinico-pathological spectrum of the same disease process. Thus, motor neurone disease is now regarded as a multi-system disease in which the motor neurones tend to be affected earliest and most severely (Ince et al. 1998a).

Gross pathological changes. The gross pathological changes of amyotrophic lateral sclerosis consist of atrophy of the cerebral precentral gyrus, and shrinkage, sclerosis, and pallor of the lateral and anterior corticospinal tracts tracts of the spinal cord. Thinning of the hypoglossal nerves and anterior spinal roots may be observed, and there is atrophy of the somatic musculature.

Lower motor neurone pathology. Motor neurone disease patients will typically have lost 50 per cent of the lower motor neurones in the limb enlargement areas of the spinal cord at autopsy (Ince 2000). Many of the remaining lower motor neurones show atrophic and basophilic changes that are likely to represent part of the spectrum of an apoptosis, programmed cell death pathway (Martin 1999). The depletion of lower motor neurones is accompanied by diffuse astrocytic gliosis in the spinal grey matter. There is relative preservation of motor neurones in the nucleus of Onufrowitz, Onuf’s nucleus, in the sacral spinal cord (Mannen et al. 1977), which innervates skeletal muscles of the pelvic floor, and in the cranial motor nuclei of the oculomotor, trochlear, and abducens nerves which control eye movements. The selective resistance of these two groups of motor neurones in motor neurone disease is unexplained.

A cardinal feature of lower motor neurone pathology in motor neurone disease is the presence of inclusion bodies within the soma and proximal dendrites. Ubiquitinated inclusions are the most frequent neuronal lesion, and are found in virtually 100 per cent of cases (Ince et al. 2003; Piao et al. 2003). These inclusions show a range of appearances including thread-like profiles; skeins (Fig. 23.3A) of varying compactness and more compact spherical bodies (Fig. 23.3C) (Ince et al. 1998a). Ubiquitin is a small highly conserved protein which becomes covalently bound to intracellular proteins targeted for disposal. The resulting tagged protein is degraded in the proteolytic channel of the 26S proteasome. Ubiquitin immunoreactivity is a prominent feature of the neuropathology of many neurodegenerative diseases, where it is found as a component of various inclusion bodies. Whereas in several other neurodegenerative diseases, the protein to which ubiquitin binds is known, for instance tau or α-synuclein ubiquitin-positive inclusions, in motor neurone disease the protein substrate for ubiquitination is not currently known with certainty, though TD43 represents an important candidate (Neumann et al. 2006).

 Inclusion bodies within spinal cord motor neurones in amyotrophic lateral sclerosis. A. Skein like ubiquitinated inclusion; B. Hyaline conglomerate neurofilament inclusion body in a patient with superoxide dismutase related familial amyotrophic lateral sclerosis; C. Compact ubiquitinated inclusion; A,C immunohistochemistry for ubiquitin; B immunohistochemistry for neurofilament: all ×40 obj. Arrows in A, B, and C indicate the relevant inclusion.
Fig. 23.3

Inclusion bodies within spinal cord motor neurones in amyotrophic lateral sclerosis. A. Skein like ubiquitinated inclusion; B. Hyaline conglomerate neurofilament inclusion body in a patient with superoxide dismutase related familial amyotrophic lateral sclerosis; C. Compact ubiquitinated inclusion; A,C immunohistochemistry for ubiquitin; B immunohistochemistry for neurofilament: all ×40 obj. Arrows in A, B, and C indicate the relevant inclusion.

Bunina bodies, first described in 1962 are eosinophilic inclusions present in the soma of lower motor neurones (Bunina 1962). They are found in 86 per cent of amyotrophic lateral sclerosis cases (Piao et al. 2003), and are specific to the disease. They have been shown to be immunoreactive to the proteinase inhibitor cystatin C, but their nature, origin, and significance remain unclear (Okamoto et al. 1993).

Neurofilament conglomerate inclusions are large inclusion bodies detectable with silver stains and with antibodies directed at both the phosphorylated and non-phosphorylated forms of heavy- and medium-chain neurofilament proteins (Ince et al. 1998b). Motor neurones containing these prominent inclusion bodies lose the normal neurofilament cytoskeleton from the remainder of the cell body. Neurofilament conglomerate inclusions are seen only infrequently in sporadic cases of motor neurone disease and are most commonly described in familial motor neurone disease caused by specific mutations in the SOD1 gene (Fig. 23.3B) (Rouleau et al. 1996; Ince et al. 1998). In normal motor neurones, neurofilament proteins within the cell body are predominantly non-phosphorylated, and there is progressive phosphorylation of neurofilaments in the axonal compartment. Several reports have documented a diffuse increase in neurofilament phosphorylation within the soma of spinal motor neurones in motor neurone disease (Munoz et al. 1988; Sobue et al. 1990). Swellings termed spheroids have been described in the axons of lower motor neurones in motor neurone disease. These are composed of abnormally orientated accumulations of neurofilaments, and are presumed to represent focal abnormalities of axonal cytoskeletal regulation, with failure of axonal transport (Ince 2000). Axonal spheroids are not however, disease specific, and are present in normal subjects, although the numbers of spheroids may be increased in motor neurone disease. Spheroids have been described in the cell body of spinal motor neurones (Hirano et al. 1967), and by immunocytochmistry appear similar to neurofilament conglomerate inclusions suggesting that the same dysregulation of cytoskeletal function may underlie both types of cellular lesion.

Upper motor neurone pathology. A major feature of the pathology of motor neurone disease is axonal loss within the descending pyramidal motor pathway, associated with secondary myelin pallor and gliosis of the corticospinal tracts. This pathology gives rise to the ‘lateral sclerosis’ within the spinal cord originally described by Charcot. Myelin pallor of the corticospinal tract is most prominent in the cervical cord and medullary pyramids, and in many cases is not demonstrable above the level of the medulla. This finding suggests that upper motor neurone changes in motor neurone disease arise from a dying back axonopathy, with distal denervation due to axonal loss preceding degeneration of the cell bodies of the upper motor neurones and loss of pyramidal cells from the cortex (Ince 2000).

In the motor cortex, pathological changes are highly variable, even in patients with well-established signs of upper motor neurone involvement clinically. Severely affected cases will usually show a reduction in the population of giant pyramidal neurones, Betz cells, in the motor cortex, either due to loss of these neurones, or a reduction in their size so that they are indistinguishable from smaller neighbouring pyramidal cells. Intracellular inclusions, including ubiquitinated inclusions, have not been convincingly described in Betz cells. The motor cortex may show astrocytic gliosis of varying severity in the grey matter and underlying subcortical white matter. Evidence of microglial activation, can be detected with immunocyotchemical markers in the corticospinal tract within the brain and spinal cord (Troost et al. 1990; Ince et al. 2003).

Sensory and cerebellar pathways. Despite the paucity of sensory signs clinically, the ascending sensory pathways of the dorsal column are commonly affected in amyotrophic lateral sclerosis. In the Japanese literature, the prevalence of dorsal column pallor in familial disease is emphasized, with the concept of ‘familial amyotrophic lateral sclerosis with posterior column involvement’. However, more recent work suggests that this change is detectable at autopsy in up to 50 per cent of all sporadic amyotrophic lateral sclerosis cases (Ince et al. 2003). There is also evidence of degenerative changes in peripheral sensory nerves, and loss of large afferent nerve fibres (Kawamura et al. 1981). Cerebellar involvement in amyotrophic lateral sclerosis is not usually recognized clinically, although late involvement of these pathways could go undetected in the presence of severe weakness. Pathologically, degeneration of the spinocerebellar pathway, reflected by cell loss from the thoracic nucleus of Clarke, and by pallor of ascending spinocerebellar pathways, is a relatively frequent finding in sporadic motor neurone disease (Brownell et al. 1970).

Muscle pathology. Loss of lower motor neurones result in denervation and atrophy of skeletal muscle. Histologically denervated muscle shows clusters of angular atrophic fibres and fibre-type grouping which results from serial denervation and reinnervation arising from collateral sprouting of axons of surviving motor neurones within muscle (Ince 2000).

Primary lateral sclerosis and progressive muscular atrophy. The pathological features of several patients with primary lateral sclerosis have been described (Pringle et al. 1992; Watanabe et al. 1997). The most prominent features described are atrophy of the prefrontal gyrus, with loss of Betz cell somata, and pallor and atrophy of the lateral corticospinal tracts. In cases where ubiquitin immunochemistry was undertaken, classical ubiquitinated inclusions were seen in a small number of lower motor neurones, and gliosis of the spinal ventral horns has also been described (Pringle et al. 1992; Watanabe et al. 1997). Ubiquitinated inclusion bodies in the cerebral cortex in a distribution typically found in amyotrophic lateral sclerosis-dementia cases has also been described (Kawashima et al. 1998), suggesting that primary lateral sclerosis may merge into the spectrum of amyotrophic lateral sclerosis dementia.

A pathological study of 14 patients with the clinical phenotype of progressive muscular atrophy demonstrated typical ubiquitinated inclusions in the spinal cord or bulbar motor neurone groups in most cases (Ince et al. 2003). Interestingly, 50 per cent of this series were also shown to have evidence of corticospinal tract involvement demonstrated by immunostaining for active microglia and macrophages.

The pathological findings of occasional abnormalities of the lower motor neurones in primary lateral sclerosis, and of corticospinal tract involvement in progressive muscular atrophy support the clinical evidence of overlap between these conditions and amyotrophic lateral sclerosis (Rowland 1999). Ubiquitinated inclusions are the characteristic pathological feature of amyotrophic lateral sclerosis, primary lateral sclerosis, and progressive muscular atrophy suggesting that these disorders share common pathophysiological mechanisms (Mackenzie and Feldman 2005). Primary lateral sclerosis and progressive muscular atrophy should be considered part of the spectrum of amyotrophic lateral sclerosis.

Pathological features of prolonged disease. Patients whose survival is prolonged by invasive ventilation may develop clinical and pathological features of involvement of less vulnerable motor neurone groups and eventually widespread involvement of the central nervous system (Hayashi and Kato 1989; Mizutami et al. 1990; Sasaki et al. 1992). These patients may lose voluntary control of the ocular muscles, and become totally ‘locked-in’. They also lose control of external sphincters, and develop decubitus ulcers, features not typically seen in motor neurone disease. Electroencephalograms performed at later stages of disease show diffuse slowing (Hayashi et al. 1989) and MR imaging demonstrates progressive cerebral atrophy, including the frontal and temporal lobes, precentral gyrus, postcentral gyrus, anterior cingulate gyrus, and corpus callosum (Kato et al. 1993).

Pathology of cognitive impairment. Patients with motor neurone disease and dementia have both the characteristic motor system ubiquitinated inclusions, together with cerebral pathology, which consists of small globular ubiquitinated inclusions within the dentate granule cells (Fig. 23.4), and a variable component of neocortical ubiquitinated neurites and small neuronal ubiquitinated inclusions (Munoz et al. 2003; Mackenzie and Feldman 2003). Other limbic structures, including the amygdala and parahippocampal gyrus may also show ubiquitinated inclusion pathology. Autopsy series indicate that 20–50 per cent of non-demented motor neurone disease patients have similar cerebral pathology, although the severity and distribution tends to be less extensive (Wilson et al. 2001; Mackenzie et al. 2003; Mackenzie and Feldman 2005). The degree of overlap in pathological findings, neuropsychological deficits, and imaging studies in classical motor neurone disease without dementia, motor neurone disease inclusion dementia, and amyotrophic lateral sclerosis-fronto-temporal dementia suggest that they represent a spectrum of clinical disease with a common pathological substrate.

 Ubiquitinated inclusions in the dentate granule cells of the hippocampus in a patient with amyotrophic lateral sclerosis dementia. Immunohistochemsitry for ubiquitin: ×20 obj.
Fig. 23.4

Ubiquitinated inclusions in the dentate granule cells of the hippocampus in a patient with amyotrophic lateral sclerosis dementia. Immunohistochemsitry for ubiquitin: ×20 obj.

The primary pathogenetic processes underlying amyotrophic lateral sclerosis are multifactorial and the precise mechanisms underlying selective cell death in the disease are at present incompletely understood. Current understanding of the neurodegenerative process in amyotrophic lateral sclerosis suggests that there may be a complex interplay between multiple mechanisms including genetic factors, oxidative stress, excitotoxicity, and protein aggregation as well as damage to critical cellular processes, including axonal transport and organelles such as mitochondria (Bruijn and Cleveland 1996; Bruijn et al. 2004; Shaw 2005). Recently there has been growing interest in the role played in motor neurone injury by neighbouring non-neuronal glial cells and in dysfunction of particular molecular signalling pathways. The relative importance of these different pathways may well vary in different subgroups of patients, and a very important task for the future is to further define the subgroups of amyotrophic lateral sclerosis. Evidence has also accumulated that the final process of motor neurone death is likely to occur via a caspase-dependent programmed cell death pathway resembling apoptosis.

Patients with amyotrophic lateral sclerosis report a family history of the disease in about 5–10 per cent of cases. The most common pattern of inheritance is autosomal dominant, with complete penetrance, although recessive or X-linked inheritance occurs in some pedigrees. Linkage studies in amyotrophic lateral sclerosis are rendered difficult by age-dependent onset in adults, short disease duration, heterogeneity of presentation, and misdiagnosis. Linkage to 12 different chromosomal loci has been established in familial motor neurone disease, and for 6 of these, the underlying genetic defect has been identified (Table 23.4).

Table 23.4
Genetics of familial amyotrophic lateral sclerosis (ALS) and genetic association factors in sporadic amyotrophic lateral sclerosis
Age of onsetInheritanceChromosomeGene

FAMILIAL ALS

 ALS1

Adult

Dominant

21q22.1

SOD1

 ALS2

Juvenile

Recessive

2q33

ALSIN

 ALS3

Adult

Dominant

18q21

 ALS4

Juvenile

Dominant

9q34

Senataxin

 ALS5

Juvenile

Recessive

15q15.1-q21.1

 ALS6

Adult

Dominant

16q12

 ALS7

Adult

Dominant

20ptel-p13

 ALS8

Adult

Dominant

20q13.33

VAPB

 ALS-X

Adult

Dominant

Xp11-q12

ALS-DEMENTIA

 ALS-FTD

Adult

Dominant

9q21-22

 ALS-FTD

Adult

Dominant

9p21.3-13.3

 ALS-PD-dementia

Adult

Dominant

17q21-22

MAPT

 MND-ID

Adult

Dominant

17q21-22

not tau related

 DLDH and ALS

Adult

Dominant

3

CHMP2B

Age of onsetInheritanceChromosomeGene

FAMILIAL ALS

 ALS1

Adult

Dominant

21q22.1

SOD1

 ALS2

Juvenile

Recessive

2q33

ALSIN

 ALS3

Adult

Dominant

18q21

 ALS4

Juvenile

Dominant

9q34

Senataxin

 ALS5

Juvenile

Recessive

15q15.1-q21.1

 ALS6

Adult

Dominant

16q12

 ALS7

Adult

Dominant

20ptel-p13

 ALS8

Adult

Dominant

20q13.33

VAPB

 ALS-X

Adult

Dominant

Xp11-q12

ALS-DEMENTIA

 ALS-FTD

Adult

Dominant

9q21-22

 ALS-FTD

Adult

Dominant

9p21.3-13.3

 ALS-PD-dementia

Adult

Dominant

17q21-22

MAPT

 MND-ID

Adult

Dominant

17q21-22

not tau related

 DLDH and ALS

Adult

Dominant

3

CHMP2B

SPORADIC ALS GENETIC ASSOCIATIONSVariantChromosomeGene

6 del / 2 ins

22q12

Neurofilament

heavy KSP region

1 del

Mitochondria

Cytochrome c oxidase

Point mutation

Mitochondria

tRNA (Ile)

1 del

14q11-q12

AP endonuclease

Splicing

11p13

EAAT2

Promotor SNPs

6p12

VEGF

Copy number

5q13

SMN

E4 isoform

19q13

Apo E

Point mutations

14q11.2

Angiogenin

Point mutations

3p12.1

CHMP2B

SPORADIC ALS GENETIC ASSOCIATIONSVariantChromosomeGene

6 del / 2 ins

22q12

Neurofilament

heavy KSP region

1 del

Mitochondria

Cytochrome c oxidase

Point mutation

Mitochondria

tRNA (Ile)

1 del

14q11-q12

AP endonuclease

Splicing

11p13

EAAT2

Promotor SNPs

6p12

VEGF

Copy number

5q13

SMN

E4 isoform

19q13

Apo E

Point mutations

14q11.2

Angiogenin

Point mutations

3p12.1

CHMP2B

ALS 1: Copper–zinc superoxide dismutase, SOD1. Familial motor neurone disease with adult onset is clinically indistinguishable from sporadic amyotrophic lateral sclerosis in individual cases. Twenty per cent of families with autosomal dominant motor neurone disease show mutations in the gene on chromosome 21q22.1 which encodes the free radical scavenging enzyme superoxide dismutase, referred to as SOD1 (Rosen et al. 1993). More than 100 mutations have now been identified, the majority of which are missense mutations. SOD1 is a ubiquitously expressed metalloenzyme whose major function is to convert intracellular superoxide free radicals to hydrogen peroxide. SOD1 is an abundant protein in the central nervous system, accounting for about 1 per cent of brain protein. SOD1 was initially thought to be confined to the cytosolic compartment of cells but it is now recognized that a small proportion of the protein is located in the intermembrane space of mitochondria (Okado-Matsumoto and Fridovich 2001). The reasons why motor neurones are especially vulnerable to injury in the presence of SOD1 mutations, are not yet clear. Despite 13 years of intensive research effort, the pathways leading to the cell death of motor neurones in the presence of SOD1 mutations have not been fully identified, although there is a convincing body of evidence that the mutant SOD1 protein exerts its detrimental effects through a toxic gain of function rather than a loss of function. Most of our current level of understanding of disease mechanisms in amyotrophic lateral sclerosis has come from the study of the effects of SOD1 mutations but, even in this defined genetic subgroups of disease, the pathways to neurodegeneration appear to be complex and multifactorial.

There is considerable variation in disease phenotype in terms of age of onset and rate of disease progression in human SOD1 related motor neurone disease. It is apparent that the clinical phenotype must be modified by other genetic and/or environmental factors. There has been much interest in the D90A SOD1 mutation, which has a dominant inheritance in some genetic backgrounds, but is recessively inherited with two mutated copies of the gene required to cause disease, in Scandinavian populations, implying a co-inherited protective factor (Andersen et al. 1996).

Mutant SOD1 transgenic mice have been genetically engineered and develop a disease which clinically and pathologically resembles human motor neurone disease. The most extensively studied are SOD1 G93A, SOD1 G37R, and SOD1 G85R (Bruijn et al. 1996). Transgenic rats, carrying G93A or H46R SOD1 also develop a motor neurone disease phenotype (Nagai et al. 2001). In addition cellular models of SOD1 related motor neurone disease have been generated which have helped to elucidate cellular mechanisms of disease (Pasinelli et al. 1998; Cookson et al. 2002). The toxic gain of function of mutant SOD1 has not yet been fully defined, but there are several pathophysiological processes which may be involved, including oxidative stress, mitochondrial dysfunction, excitotoxicity, protein aggregation, and inflammation. These mechanisms are not mutually exclusive and it is possible that all of these factors play a role in the development of motor neurone injury.

ALS 2: Alsin. In 2001 alsin was identified as the causative gene for an autosomal recessive form of juvenile amyotrophic lateral sclerosis linked to chromosome 2q33 (Hadano et al. 2001; Yang et al. 2001). Mutations in alsin can also cause a motor neurone degenerative disorder with a predominant upper motor neurone phenotype: juvenile recessive primary lateral sclerosis; infantile onset ascending hereditary spastic paralysis (Eymard-Pierre et al. 2002); or autosomal recessive complicated hereditary spastic paraplegia (Gros-Louis et al. 2003).

ALS 2 encodes a 184 KDa protein which contains three putative guanine nucleotide exchange factor domains and is alternatively spliced to generate a short and a long transcript. These factors are known to activate small GTPase proteins by stimulating the release of GDP in exchange for GTP. Given the conserved guanine nucleotide exchange factor domains of ALS2, it is predicted to function as an activator of particular small GTPases. The small GTPases control a range of important cellular processes and function as binary switches—alternating between inactive GDP-bound and active GTP-bound states. The alsin protein is widely expressed, but enriched within the central nervous system, where it is localized to the cytoplasmic face of endosomal membranes (Yamanaka et al. 2003). The functions of alsin are still being investigated, but to date it has been shown to function as an activator of the small GTPase protein Rab5 (Otomo et al. 2003; Kunita et al. 2004). This implies that alsin is important in endosomal dynamics and the working hypothesis is that alsin normally regulates trafficking of signalling molecules important for proper development and/or maintenance of health of motor neurones. ALS2 knockout mice have been generated but the motor system phenotype so far arising appears to be very mild (Kris et al. 2003).

ALS 4: Senataxin The ALS4 locus linked to chromosome 9q34 was originally identified in a single large pedigree with juvenile onset, autosomal dominant amyotrophic lateral sclerosis. The disease course in this family was indolent and did not reduce life expectancy. Three different missense mutations, L3095, R2136H, and T3I, in three families with this subtype of motor neurone disease were identified (Chen et al. 2004). The SETX gene encodes senataxin, a large 302.8KD protein of unknown function. Much of the protein has no homology with other known proteins but there is one DNA/RNA helicase domain. DNA/RNA helicase proteins are known to have roles in processes such as repair, replication, recombination or transcription of DNA and RNA processing, RNA transcript stability, and the initiation of translation. Recessive loss of function mutations in SETX are associated with ataxia-oculomotor apraxia type 2 (Moreira et al. 2004). It is predicted that the different phenotype of dominantly inherited ALS4 is likely to be caused by a toxic gain of function of the mutated senataxin protein.

ALS 8: VAPB. Nishimura and co-workers described a novel missense mutation, P565, in the VAPB gene for vesicle-associated membrane protein/synaptobrevin associated membrane protein, located on chromosome 20q13.3, in a Brazilian family with ALS8, an autosomal dominant slowly progressive disorder characterized by fasciculation, cramps, and postural tremor (Nishimura et al. 2004). They subsequently found the same mutation in six further families with different clinical phenotypes, including late onset spinal muscular atrophy and classical rapidly progressive amyotrophic lateral sclerosis. Vesicle associated proteins are intracellular membrane proteins that can associate with microtubules and have been shown to function in membrane transport. The VAPB protein has three identifiable structural domains. The first 150 residues form an MSP domain conserved between all members of this protein family; the central region contains an amphipathic helical structure predicted to form a coiled/coil protein–protein interaction motif and at the carboxy terminus is a hydrophobic region that acts as a membrane anchor. Preliminary cell biological studies have indicated that the wild-type VAPB protein localizes predominantly to the endoplasmic reticulum. The P56S mutation disrupts the subcellular distribution and induces the formation of intracellular protein aggregates (Nishimura et al. 2004).

Dynactin mutation. A mutation, G595, substitution in the gene encoding the P150 subunit of dynactin, DCTN1, has been identified in a single family with a slowly progressive lower motor neurone degenerative disorder (Puls et al. 2003). The described family had an unusual phenotype, presenting in early adulthood with respiratory difficulties due to vocal cord paralysis, progressive facial weakness, and weakness and atrophy of the hands and later development of neurogenic changes distally in the lower limbs. Mutations of the p150 subunit of DCTN1 were subsequently identified in patients with familial amyotrophic lateral sclerosis and frontotemporal dementia, and one case of apparently sporadic amyotrophic lateral sclerosis (Munch et al. 2004; Munch et al. 2005).

The dynactin–protein complex is required for dynein mediated retrograde axonal transport of vesicles and organelles along the microtubule system. The amino acid change caused by the mutation is predicted to distort the folding of the microtubule binding domain of dynactin. Overexpression of the P50 subunit of dynactin has been shown to disrupt the function of this protein complex and causes late onset progressive motor neurone degeneration in genetically engineered mice (La Monte et al. 2002).

Other amyotrophic lateral sclerosis loci. The genes for several other subtypes of amyotrophic lateral sclerosis remain to be identified. Three separate families have shown linkage to chromosome 16. Amyotrophic lateral sclerosis with fronto-temporal dementia has been mapped to a 17-cM interval chromosome 9q21 (Hosler et al. 2000; Ostojic et al. 2003), and one Swedish family with a similar phenotype without linkage to the chromosome 9 locus has recently been identified, suggesting genetic heterogeneity for this subtype of disease. Motor neurone degeneration may occur in patients with fronto-temporal dementia and Parkinson’s disease associated with mutations in the microtubule associated protein tau MAPT (Clark et al. 1998). The mutant tau protein forms filamentous inclusions and insoluble aggregates that are associated with neurodegeneration. Some patients with familial fronto- temporal dementia, Parkinsonism, and amyotrophic lateral sclerosis do not have identified mutations in tau suggesting that further genes causing this triad of features remain to be identified (Kowalska et al. 2003).

Possible genetic risk factors in sporadic amyotrophic lateral sclerosis. There have been reports of genetic variants found in individuals with apparently sporadic disease (summarized in Table 23.4) (Kunst 2004; Shaw 2005).

Cellular injury by free radical species is a major potential cause of the age-related deterioration in neuronal function which occurs in neurodegenerative diseases. There has been particular interest in the role of oxidative stress in amyotrophic lateral sclerosis given that SOD1 mutations, which encode a key cellular anti-oxidant defence protein, underlie approximately 20 per cent of familial cases. Studies of CSF and human post-mortem central nervous system tissue have demonstrated the presence of biochemical changes which represent the effects of oxidative stress and these changes are more pronounced in amyotrophic lateral sclerosis cases compared to controls (Shaw et al. 1995b; Ferrante et al. 1997; Smith et al. 1998; Tohgi et al. 1999). Fibroblasts cultured from the skin of patients with both familial and sporadic amyotrophic lateral sclerosis show increased sensitivity to oxidative insults compared to those from control cases (Aguirre et al. 1998).

In relation to the toxic gain of function of the mutant superoxide dismutase 1 protein, oxidative damage and/or metal mishandling have been strongly implicated. The main hypotheses have been that mutations alter the structure of this protein, allowing greater access of abnormal substrates to the active copper site of the dimeric enzyme, resulting in the production of damaging free radical species including peroxynitrite and hydroxyl radicals. Nitration of tyrosine residues on cellular proteins by peroxynitrite can have damaging consequences (Beckman et al. 1993). Some SOD1 mutations render the protein more likely to form a zinc deficient variant (Crow et al. 1997), which in turn makes the copper site more accessible to abnormal substrates. In vitro studies have demonstrated that zinc deficient superoxide dismutase 1 causes peroxynitrite dependent cell death (Estevez et al. 1999). However, a body of experimental work has raised questions as to whether the toxicity of mutant superoxide dismutase 1 can be explained by copper- dependent oxidative mechanisms. For example, superoxide dismutase 1 that has been manipulated not to bind copper by mutating the four histidine residues for copper binding still causes motor neurone disease in transgenic mice. Also, knock out of the gene encoding the copper chaperone protein normally required for insertion of copper into the enzyme, has no effect on the disease phenotype in SOD1 transgenic mice (Subramaniam et al. 2002). It seems plausible that mutant superoxide dismutase 1 may induce oxidative stress by a mechanism beyond its own catalytic activity and transcriptional repression of anti-oxidant response genes under the control of the transcription factor NRF2 is one potential pathway of interest (Kirby et al. 2005).

Glutamate is the major excitatory transmitter in the human central nervous system and there is great complexity in the molecular structure of the repertoire of receptors for this neurotransmitter system. Excitotoxicity is the term coined for neuronal injury induced by excessive stimulation of glutamate receptors, by mechanisms which include derangement of intracellular calcium homeostasis, and excessive free radical production. Motor neurones appear particularly susceptible to toxicity via activation of cell surface AMPA receptors (Carriedo et al. 1996). Glutamatergic toxicity has been implicated as a contributory factor to motor neurone injury in amyotrophic lateral sclerosis (Heath and Shaw 2005). The key findings are that the expression and function of the major glial glutamate re-uptake transporter protein EAAT2 may be impaired in the central nervous system of motor neurone disease patients and that CSF, and therefore central nervous system extracellular fluid, levels of glutamate appear to be abnormally elevated at least in a proportion of motor neurone disease patients (Rothstein et al. 1995; Shaw et al. 1995a; Fray et al. 1998; Spreux-Varoquaux et al. 2002). Excitotoxicity has provided a potential mechanistic link between SOD1 mutant mediated motor neurone disease and the sporadic form of the disease. The presence of mutant superoxide dismutase 1 increases the sensitivity of motor neurones to glutamate toxicity (Kruman et al. 1999); causes alteration in AMPA receptor subunit expression (Spalloni et al. 2004); as well as reduced expression of the major glutamate re-uptake transporter EAAT2 (Bendotti et al. 2001). Whether as a primary or a propagating process, it appears that glutamate toxicity plays a contributory role to the injury of motor neurones in amyotrophic lateral sclerosis. This is supported by the finding that anti-glutamate therapy with riluzole has some effect, albeit modest, in prolonging survival in human amyotrophic lateral sclerosis patients, and in SOD1 mutant mouse models (Lacomblez et al. 1996; Gurney et al. 1996).

Mitochondria serve multiple important intracellular functions including generation of intracellular ATP, buffering of intracellular calcium, generation of intracellular free radicals, and involvement in the initiation of apoptotic cell death. Age-related deterioration in mitochondrial function may contribute to the development of late-onset neurodegenerative diseases. There is a body of evidence emerging from investigation of human material and cellular and animal models indicating that mitochondrial dysfunction may contribute to motor neurone injury in amyotrophic lateral sclerosis and this has been reviewed (Beal 2000; Menzies et al. 2002b).

The key evidence for mitochondrial dysfunction in human amyotrophic lateral sclerosis includes:

alteration in the morphology of mitochondria in hepatocytes, muscle and motor neurones;

increased mitochondrial volume and calcium levels within motor axon terminals in muscle biopsies from sporadic amyotrophic lateral sclerosis cases (Siklos et al. 1996);

reduced complex IV activity in spinal motor neurons in sporadic amyotrophic lateral sclerosis (Borthwick et al. 1999);

high frequency of mitochondrial DNA mutations in motor cortex tissue in sporadic amyotrophic lateral sclerosis (Dhaliwal et al. 2000);

Multiple mutations and decreased mitochondrial DNA in muscle and spinal cord in sporadic motor neurone disease (Wiedemann et al. 2002); and

amyotrophic lateral sclerosis like phenotypes in several patients with genetic mutations affecting mitochondrial proteins (Comi et al. 1998; Borthwick et al. 2006).

In cellular models of superoxide dismutase 1 related motor neurone disease, expression of a G93A SOD1 mutant results in the development of abnormally swollen mitochondria which display impaired activity of complexes II and IV of the mitochondrial respiratory chain, impaired cellular bioenergetic status, and alteration in the mitochondrial proteome (Menzies et al. 2002a; Wood-Allum et al. 2006). Molecular targeting of mutant superoxide dismutase 1 to the mitochondria but not to the nucleus or endoplasmic reticulum leads to activation of the apoptosis cascade and cell death (Takeuchi et al. 2002). In some strains of mutant SOD1 transgenic mice mitochondrial vacuolation within motor neurones is an early feature of the pathology (Wong et al. 1995). Whereas superoxide dismutase 1 was previously considered to be an exclusively cytosolic protein, it is now recognized also to reside in the intermembrane space of mitochondria. Superoxide dismutase 1 has been shown to accumulate in vacuolated mitochondria in mutant SOD1 mice. It has been demonstrated that the activities of several complexes of the mitochondrial respiratory chain are reduced prior to disease onset and that these changes increase with age. Oxidative damage to mitochondrial protein and lipids and decreased ATP synthesis have been reported at the onset of the murine disease. Translocation of cytochrome C, an initiator of apoptosis, from the mitochondria to the cytosol has been demonstrated during disease progression in the mice. Recent studies have reported that mutant superoxide dismutase 1 is selectively and aberrantly recruited to the cytoplasmic face of mitochondria in spinal cord tissue from mutant SOD1 transgenic mice and that the anti-apoptotic protein Bcl2 may be entrapped within large protein aggregates of superoxide dismutase 1 within spinal cord tissue, which may result in reduced availability of this protein to regulate apoptosis (Liu et al. 2004; Pasinelli et al. 2004).

Therapeutic effects of compounds which modulate mitochondrial function have begun to be investigated in SOD1 transgenic mouse models. Creatine buffers energy levels within the cell, maintains ATP levels, and stabilizes mitochondrial creatine kinase which inhibits opening of the mitochondrial permeability transition pore. Administration of creatine to G93A transgenic mice improved motor function and extended survival in a dose-dependent manner, as well as causing a decrease in biochemical indices of oxidative damage in the spinal cord (Klivenyi et al. 1999). Minocycline, a tetracycline derivative which inhibits microglial activation and blocks release of cytochrome c from mitochondria, also slows disease in mutant SOD1 mice (Zhu et al. 2002).

Motor neurones, which in the human nervous system may have axons up to 1 m in length, are highly reliant on an efficient intracellular transport system with anterograde and retrograde components. It is interesting that in SOD1 mutant mice, axonal transport is demonstrably impaired several months before clinical disease onset (Wiliamson et al. 1999). The kinesin complex of proteins are important molecular motors for anterograde axonal transport on the microtubule system. Mutations of genes encoding several kinesin proteins have been shown to cause several types of motor neurone degeneration including a hereditary spastic paraplegia, SPG10, and type 2A Charcot–Marie–Tooth disease, though have not yet been associated with amyotrophic lateral sclerosis. The dynein–dynactin complex is the important motor for retrograde transport on the microtubule system, returning components such as multivesicular bodies and neurotrophic factors back to the perikaryon. Mutations in dynein and the dynactin complex which is an activator of cytoplasmic dynein, cause progressive motor neurone disease in mice (Lamonte et al. 2002; Hafezparast et al. 2003). Mutations in the P150 subunit of dynactin may cause a form of motor neurone disease in human subjects (Puls et al. 2003).

Neurofilament proteins form a major component of the cytoskeleton of neurones and important functions include maintenance of cell shape and axonal calibre, as well as axonal transport. Neurofilament subunits are assembled in the motor neurone cell body, and transported down the axon by slow axonal transport. Accumulation and abnormal assembly of neurofilaments are common pathological hallmarks of amyotrophic lateral sclerosis. Ubiquitinated inclusions with compact or Lewy body like morphology within surviving motor neurones in amyotrophic lateral sclerosis may show immunoreactivity for neurofilament epitopes. In some cases of SOD1 mutation related amyotrophic lateral sclerosis, large argyrophilic neurofilament conglomerate inclusions have been observed in the cell bodies and axons of motor neurones (Ince et al. 1998). Approximately 1 per cent of sporadic amyotrophic lateral sclerosis cases have deletions of insertions in the KSP repeat region of the neurofilament heavy chain gene, NFH (Figlewicz et al. 1994; Tomkins et al. 1998). Pathological changes within motor neurones develop in mice overexpressing NF-light or -heavy subunits, or in mice expressing mutations in the NFL gene. Transgenic mice which carry SOD1 mutations, also show alterations in neurofilament organization, with the development of neurofilament spheroids, as well as reduced neurofilament protein and decreased transport rate in the ventral root axons. Genetic manipulations to alter the expression of neurofilament proteins have been shown to alter the disease course in SOD1 transgenic mice. Increased expression of NFH, resulting in trapping most neurofilaments within the cell body, robustly improves the disease course, by as much as 6 months in mutant SOD1 mice (Couillard-Despres et al. 1998).

Misfolding of proteins with the formation of intracellular aggregates is a key feature of multiple neurodegenerative diseases. There is continuing debate as to whether such aggregated proteins play an important role in disease pathogenesis, whether they represent harmless by-standers, or whether they could be beneficial to the cell by sequestration of toxic abnormal proteins. In the SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis, the mutant superoxide dismutase 1 protein forms conspicuous cytoplasmic inclusions in motor neurones and sometimes in astrocytes, which develop before the onset of motor dysfunction. Several hypotheses have been put forward to explain how mutant superoxide dismutase 1 aggregates could generate cellular toxicity:

Sequestration of other proteins required for normal motor neurone function. Several additional proteins have been shown to be present in superoxide dismutase 1 aggregates including: CCS, the copper chaperone for SOD1, ubiquitin neurofilaments, glial fibrillary acidic protein, two neuronal glutamate transporters, BCl2 and proteins involved in chaperone and proteosome functions.

By repeatedly misfolding, the superoxide dismutase 1 aggregates may reduce the availability of chaperone proteins required for the folding and function of other essential intracellular proteins.

The superoxide dismutase 1 mutant protein aggregates may reduce proteasome activity needed for normal protein turnover.

Inhibition of the function of specific organelles such as mitochondria by aggregation on or within these organelles.

Overexpression of chaperone proteins has been shown to reduce mutant superoxide dismutase 1 aggregation and enhances the survival and function of motor neurones in culture (Takeuchi et al. 2002). In addition, arimoclomol, a drug which up-regulates the expression of heat shock proteins increases the life span of G93A SOD1 mice by 22 per cent (Kieran et al. 2004). Clearly protein aggregates, which can be identified by ubiquitin immunostaining, are a feature of sporadic as well as familial motor neurone disease. Superoxide dismutase 1 containing aggregates are not a characteristic feature of sporadic motor neurone disease and determining the nature of the protein inclusions in the sporadic disease is a very important research goal. A recent report has identified TDP-43, the TAR-DNA-binding protein of 43kDa, as a component of ubiquitinated inclusions in ALS (Neumann et al. 2006). TDP-43 is known to be a nuclear factor which plays a role in the regulation of transcription and alternative splicing. Further investigation is required to determine the role played by this protein in the pathogenesis of amyotrophic lateral sclerosis.

There has been much recent interest in the possibility that non-neuronal cells, including activated microglia and astrocytes, may contribute to the pathogenesis and/or propagation of the disease process in amyotrophic lateral sclerosis. Several studies in genetically engineered mouse models have suggested that expression of mutant superoxide dismutase 1 in neurones alone is insufficient to cause motor neurone degeneration and that involvement of non-neuronal cells in the vicinity of motor neurones may be required. Chimeric mice have been produced which have both normal and mutant SOD1 expressing cells (Clement et al. 2003). Motor neurones expressing mutant superoxide dismutase 1 can escape disease if surrounded by a sufficient number of normal non-neuronal cells. Conversely normal motor neurones surrounded by mutant superoxide dismutase 1 containing non-neuronal cells, developed signs of cellular injury, with the development of ubiquitinated protein deposits. Thus, the mutant enzyme may cause neurotoxicity indirectly by perturbing the function of non-neuronal cells such as microglia (Boillee et al. 2006). Microglia play a critical role as resident immunocompetent and phagocytic cells within the central nervous system. Activation is associated with transformation to phagocytic cells capable of releasing potentially cytotoxic molecules including reactive oxygen species, nitric oxide, proteases, and pro-inflammatory cytokines (Gonzales-Scarano and Baltuch 1999). Given this, there is little doubt that activated microglia can inflict significant damage on neurones but their role is complex and they are capable of stimulating neuroprotective as well as neurotoxic effects. Proliferation of activated microglia is a prominent histological feature in the spinal ventral horn both in mutant SOD1 trangenic mice and in human amyotrophic lateral sclerosis. In the mice, microglial activation is present before the onset of significant motor neurone loss or clinical signs of disease. Various inflammatory cytokines or enzymes are up- regulated in the spinal cord or CSF of amyotrophic lateral sclerosis patients: 1L-6, 1L-1β, cyclo-oxygenase 2, and prostaglandin E2 or, in the spinal cord of mutant SOD1 mice: 1L-1β, TNF-α, cyclo-oxygenase 2, and prostaglandin E2 (Almer et al. 2002; Hensley et al. 2002; Tikka et al. 2002). Microglia appear to mediate the toxicity to neurones in culture of CSF from patients with motor neurone disease by releasing factors which enhance glutamate toxicity. Minocycline, which inhibits microglial activation ameliorates disease progression in mutant SOD1 mice (Zhu et al. 2002).

There is a tendency in amyotrophic lateral sclerosis for the disease to start focally and to spread to contiguous groups of motor neurones (Brooks et al. 1995). It would be very relevant to identify molecules that contribute to this propagation and clearly those released from activated microglia would be plausible candidates.

Apoptosis describes the controlled removal of cells by an energy-dependent cell death programme. Key molecules which regulate apoptosis include: the caspase family of proteolytic enzymes which, when activated by cleavage, orchestrate cell destruction by digesting several intracellular targets including structural and regulatory proteins; the Bcl2 family of oncoproteins where the balance and subcellular distribution between pro- and anti-apoptotic members is crucial in regulating cell survival or destruction; and the apoptosis inhibitor family of proteins which suppress apoptosis by preventing proteolytic activation of specific caspases. Several pathways triggering caspase activation have been identified including: release of pro-apoptotic factors such as cytochrome c from mitochondria; activation of cell surface ligand receptor systems of the tumour necrosis factor family including Fas-Fas ligand; and stress to the endoplasmic reticulum with activation of caspase 12.

There is evidence that motor neurones may die in amyotrophic lateral sclerosis according to a programmed cell death pathway resembling apoptosis (Guegan and Przedborski 2003; Sathasivam and Shaw 2005). Key evidence from human post-mortem studies includes:

structural morphology of degenerating motor neurones compatible with the apoptosis as well as internucleosomal DNA fragmentation detected by TUNEL staining;

increased expression of specific apoptosis related molecules for instance Ley antigen and prostate apoptosis response-4 protein in spinal cord;

alteration in the balance of expression and subcellular compartmental localization of pro- and anti-apoptotic members of the Bcl2 family in a direction favouring apoptosis; and

significant increases in the activities of caspases 1 and 3 in the spinal cord.

Investigation of cellular models of SOD1 mutation related amyotrophic lateral sclerosis has shown that motor neuronal cells expressing mutant SOD1 are more likely to die by apoptosis when oxidatively stressed (Cookson et al. 2002). In addition, under unstressed basal culture conditions, these mutant superoxide dismutase 1 containing cells appear to be ‘primed’ for cell death by expressing early molecular markers of apoptosis (Sathasivam et al. 2005). In the mutant SOD1 transgenic mouse model, there is evidence of DNA laddering, increased expression, and activation of caspase 1 and caspase 3 in the spinal cord of symptomatic mice, and alterations in the balance of key members of the Bcl2 protein family in a direction favouring apoptosis (Li et al. 2000). Cross breeding experiments between G93A SOD1 transgenic mice and mice genetically engineered to over-express anti-apopototic molecules results in amelioration of the murine disease. The administration of caspase inhibitors has a partial neuroprotective effect in cellular models (Sathasivam et al. 2005) and intraventricular administration of a broad spectrum caspase inhibitor to mutant SOD1 mice prolongs life span by approximately 20 per cent (Li et al. 2000).

One of the unsolved mysteries in neurodegenerative diseases, including amyotrophic lateral sclerosis, is the selective vulnerability of particular neuronal groups to the neurodegenerative process. Superoxide dismutase 1 is a ubiquitously distributed anti-oxidant defence protein, yet when the protein is mutated, it is the motor neurones which are most susceptible to injury. Certain cell specific features of motor neurones may predispose to age-related degeneration (Shaw and Eggett 2000; Durham et al. 2003). Key features are likely to include the cell size of motor neurones which has downstream consequences for intracellular transport, energy metabolism, and neurofilament content. The neurones vulnerable to injury in motor neurone disease have particular sensitivity to glutamatergic toxicity via AMPA receptor activation and differ from most other neuronal groups in expressing a high preponderance of calcium permeable AMPA receptors, lacking the GluR2 subunit (Williams et al. 1997). Motor neurones also have a relative lack of expression of calcium buffering proteins (Ince et al. 1993) and appear to have a high threshold for mounting a protective heat shock response. (Durham 2003) Recent studies suggest that the properties of mitochondria from the spinal cord may differ from those of mitochondria from other tissues (Sullivan et al. 2004).

The incidence of motor neurone disease reported from recent epidemiological studies ranges from 1 and 3 per 100 000, with point prevalence rates of 6–8 per 100 000 (Chancellor and Warlow 1992; Traynor et al. 1999). Most of the studies have been conducted from developed countries and relatively little is known of the incidence and prevalence in developing countries, or in specific racial or ethnic groups. Pockets of high incidence are described amongst the Chamorro indigenous population of the Western Pacific island of Guam, on the Kii peninsula of Japan and amongst the Auyu and Jakai people of Irian Jaya (Plato et al. 2003; Kuzuhara and Kokubo 2005). The explanation for the strikingly increased incidence and prevalence of motor neurone disease in these geographical foci remains uncertain. Although the prevalence remains high in Guam compared to typical populations in western countries, there has been a substantial decrease over the last half century.

Some studies have indicated that the overall age-related incidence of motor neurone disease has increased over several decades (Lilienfeld et al. 1989; Maasilta et al. 2001). However, it is unclear whether this is due to demographic factors, better ascertainment, or to changed exposure to unknown environmental risk factors. A gradual increase in the prevalence of motor neurone disease is expected given the changing age structure of the population and with the introduction of therapies which extend survival of patients.

The incidence of motor neurone disease increases with age, being very low under the age of 40 and peaking at approximately 75 years of age. The reported mean age of onset in sporadic motor neurone disease varies between 55 and 65 years in most studies, with a range varying between the third decade and the ninth decade (Jokelainen 1976; Kurtzke 1991). The mean age of onset in patients with familial motor neurone disease is about a decade earlier. Occasionally patients with classical motor neurone disease present in the second or third decade of life. Sporadic motor neurone disease is commoner in men than women, with a male/female ratio of around 1.6:1. This ratio approaches 1:1 in familial motor neurone disease. Women are relatively over-presented in older age groups, although the standardized age-related incidence is greater in elderly men. Bulbar onset is also more common in older patients, especially in older women (Haverkamp et al. 1995; Forbes et al. 2004). In most large clinic-based or population-based series, 5–10 per cent of cases are classified as familial.

The only risk factors proven to have an association with the development of motor neurone disease are gender, a positive family history, and increasing age. At the present time no environmental risk factors are regarded as of proven causative significance. Environmental factors which have been reported to increase the risk of developing motor neurone disease include trauma, physical activity, participation in athletic pursuits, dietary habits, alcohol consumption, cigarette smoking, residence in rural rather than urban areas, and working in certain occupations, for example the leather industry or electrical work. Using an evidence-based medicine approach, Armon (2003) concluded that smoking is probably associated with amyotrophic lateral sclerosis, but that the evidence in favour of other reported environmental factors was not strong. Recent studies have reported an increased risk of developing motor neurone disease in military personnel, in airline pilots, and in Italian professional football players (Horner et al. 2003; Chio et al. 2005; Weisskopf et al. 2005). Further investigation is required to substantiate these findings and to address the underlying causes of the reported occupational associations.

There are two main goals in the management of motor neurone disease. The first is the alleviation of symptoms which occur during the course of the disease to maintain quality of life. The second is the administration of neuroprotective therapy to slow the progression of motor neurone injury and neurodegeneration.

The management of motor neurone disease poses considerable ethical, logistical, and educational problems. Ethical issues are involved in aspects of management including the use of artificial methods for maintaining nutrition, ventilatory support, the use of neuroprotective drugs to slow disease progression, and the use of opiate medication in the terminal phase of the disease. The logistical and educational problems arise from the relative rarity of motor neurone disease and the fact that many health care professionals have little experience in dealing with the rapidly progressive weakness and bulbar and respiratory failure which may occur during the course of the disease. The coordinated action of multiple health care professionals within a multidisciplinary team can lessen the difficulties experienced by patients and by their families.

Symptomatic therapy aimed at alleviating the distressing symptoms which often arise during the course of motor neurone disease can do much to improve the quality of life for the patient. Detailed discussion of all of these therapies is beyond the scope of this chapter. Some of the common symptoms which may develop, and their symptomatic therapies are highlighted in Table 23.5. Many of the symptomatic therapies currently recommended by clinicians have not been assessed in rigorous controlled trials. The evidence base for some of these therapies was reviewed several years ago by an American Academy of Neurology task-force (Miller et al. 1999). A few areas of progress in the symptomatic management of motor neurone disease will be highlighted.

Table 23.5
Symptomatic therapies in amyotrophic lateral sclerosis
SYMPTOMTREATMENT

Muscle weakness and fatigue

−Physiotherapy to prevent joint stiffness and muscle contractures

 

Appliances to maintain mobility and independence such as walking aids, wheel chairs, ankle-foot orthoses, head supports, mobile arm supports bathroom aids, etc.

 

Acetylcholinesterase inhibitors (e.g. pyridostigmine) can cause a short-term improvement in fatigue in some patients, but are not used routinely

Fasciculations, cramps, spasticity

−Spasmolytic agents (baclofen, tizanidine): Dose must be carefully titrated as loss of tone can worsen mobility

−Quinine sulphate for cramps

−Low-dose diazepam for cramps or fasciculations

Sialorrhoea (drooling) and difficulty in clearing secretions

−Hyoscine transdermal patches, amitriptyline, or atropine

−Intrasalivary gland injection of botulinum toxin

−Portable suction devices

−Low-dose parotid irradiation may be considered if drug treatment is not successful.

−Carbocisteine reduces viscosity of secretions

Pseudobulbar affect

−Responds well to amitriptyline or selective serotonin reuptake inhibitors (SSRIs)

Depression and anxiety

−Tricyclic antidepressants or SSRIs

−Psychological counselling

Insomnia

− Treatment should be directed at the cause of insomnia. Common causes in motor neurone disease are respiratory insufficiency, anxiety, depression, muscle cramps, and inability to change position

−Sedatives should be administered with care in patients with respiratory compromise

Constipation

−Review medications (analgesics and anticholinergics worsen constipation) and ensure adequate fluid intake

−Bulk-forming or osmotic laxatives, glycerol suppositories

Musculoskeletal pain

−Non-steroidal anti-inflammatory agents and physiotherapy

−More potent analgesic agents may be required in the later stages

Dysarthria

− Simple strategies to improve communication can be taught by a speech therapist. When these become ineffective, a variety of communcation aids are available, such as a light-writer

Dysphagia

−Attention to food consistency, nutritional supplements.

−Gastrostomy tube insertion—radiologically guided or percutaneous endoscopic gastrostomy

Dyspnoea

−Influenza prevention vaccinations

−Antibiotics for chest infection

−Attention to sleeping position

−Sublingual lorazepam for choking, stridor

−Non-invasive ventilation

−In some countries tracheostomy and invasive ventilation may be considered

−Cough assist devices

Terminal care

−Assistance from palliative care team

−Analgesics and anxiolytic agents in incremental doses as necessary to relieve distress.

SYMPTOMTREATMENT

Muscle weakness and fatigue

−Physiotherapy to prevent joint stiffness and muscle contractures

 

Appliances to maintain mobility and independence such as walking aids, wheel chairs, ankle-foot orthoses, head supports, mobile arm supports bathroom aids, etc.

 

Acetylcholinesterase inhibitors (e.g. pyridostigmine) can cause a short-term improvement in fatigue in some patients, but are not used routinely

Fasciculations, cramps, spasticity

−Spasmolytic agents (baclofen, tizanidine): Dose must be carefully titrated as loss of tone can worsen mobility

−Quinine sulphate for cramps

−Low-dose diazepam for cramps or fasciculations

Sialorrhoea (drooling) and difficulty in clearing secretions

−Hyoscine transdermal patches, amitriptyline, or atropine

−Intrasalivary gland injection of botulinum toxin

−Portable suction devices

−Low-dose parotid irradiation may be considered if drug treatment is not successful.

−Carbocisteine reduces viscosity of secretions

Pseudobulbar affect

−Responds well to amitriptyline or selective serotonin reuptake inhibitors (SSRIs)

Depression and anxiety

−Tricyclic antidepressants or SSRIs

−Psychological counselling

Insomnia

− Treatment should be directed at the cause of insomnia. Common causes in motor neurone disease are respiratory insufficiency, anxiety, depression, muscle cramps, and inability to change position

−Sedatives should be administered with care in patients with respiratory compromise

Constipation

−Review medications (analgesics and anticholinergics worsen constipation) and ensure adequate fluid intake

−Bulk-forming or osmotic laxatives, glycerol suppositories

Musculoskeletal pain

−Non-steroidal anti-inflammatory agents and physiotherapy

−More potent analgesic agents may be required in the later stages

Dysarthria

− Simple strategies to improve communication can be taught by a speech therapist. When these become ineffective, a variety of communcation aids are available, such as a light-writer

Dysphagia

−Attention to food consistency, nutritional supplements.

−Gastrostomy tube insertion—radiologically guided or percutaneous endoscopic gastrostomy

Dyspnoea

−Influenza prevention vaccinations

−Antibiotics for chest infection

−Attention to sleeping position

−Sublingual lorazepam for choking, stridor

−Non-invasive ventilation

−In some countries tracheostomy and invasive ventilation may be considered

−Cough assist devices

Terminal care

−Assistance from palliative care team

−Analgesics and anxiolytic agents in incremental doses as necessary to relieve distress.

Specialist multidisciplinary clinics. There is an increasing tendency in the United Kingdom and worldwide for patients with motor neurone disease to be managed in specialist clinics. This allows the coordination of an experienced multi-disciplinary team, that includes the neurologist, specialist nursing staff, physiotherapist, occupational therapist, speech therapist, dietician, social worker, and orthotist. Input may also be required from other specialist teams including respiratory medicine, gastroenterology, and palliative care. Considerable support is also provided by patient associations such as the Motor Neurone Disease Association.

Nutritional management. Weight loss is universal in motor neurone disease patients and may be due to dysphagia, loss of muscle mass, or anorexia. Weight loss, malnutrition, and dehydration can aggravate muscle weakness and shorten lifespan, whilst frequent choking spells can make mealtimes intolerable. Malnutrition is an independent prognostic factor for survival in amyotrophic lateral sclerosis with an almost 8-fold increased risk of death in patients who are malnourished (Desport et al. 1999). The management of the nutritional status of motor neurone disease patients has improved in recent years. When the patient first begins to develop swallowing problems a few simple measures can be helpful. Dysphagia may be increased by anxiety and the social embarrassment resulting from slowness in eating, dribbling, and choking. Patients should be encouraged to eat in as relaxed and comfortable an environment as possible. Sucking ice before meals may decrease choking spells. Attention to food consistency is important, and the family should receive advice from a dietician. If the patient is continuing to lose weight, then nutritional supplements of liquid or semi-solid consistency may be helpful and a range of these should be tried to ascertain which preparations are most palatable for the individual patient. More active therapeutic intervention for the dysphagia should be considered when the following problems are apparent:

continuing weight loss of more than 10–20 per cent of the normal body weight despite the above measures;

dehydration;

aspiration with resultant respiratory infection; and

meal times have become too prolonged and tiring or intolerable due to frequent choking spells.

For long-term enteral feeding percutaneous endoscopic gastrostomy, or PEG, is the procedure of choice. The placement of a percutaneous endoscopic gastrostomy is a relatively straight-forward procedure which can be performed under a local anaesthetic. After placement many patients report great relief and increased well-being, though as yet no large scale quality of life studies have been conducted. Percutaneous endoscopic gastrostomy feeding in motor neurone disease has been the subject of a recent Cochrane review (Langmore et al. 2006), and represents one of the major advances in symptomatic care for patients, leading to weight stabilization and adequate nutrional and fluid intake, although a survival benefit has yet not been convincingly shown. The need for percutaneous endoscopic gastrostomy feeding should be anticipated as the risks of the procedure are higher once the patient’s forced vital capacity falls below 50 per cent (Miller et al. 1999). In some patients, technical difficulties may be experienced in the insertion of a percutaneous endoscopic gastrostomy tube and in this situation, a radiologically guided method may be used (Thornton et al. 2002). In patients with a low vital capacity, the use of non-invasive positive pressure ventilation during percutaneous endoscopic gastrostomy insertion has been shown to improve tolerance and safety of the procedure (Gregory et al. 2002). A full strength feeding regimen providing 1500–2000 calories per day can be introduced over 48 h following feeding tube insertion. Feeding can be managed by syringe boluses at normal meal-times, or by continuous infusion by pump which can be given overnight. Some patients defer consenting to undergo percutaneous endoscopic gastrostomy insertion until an advanced disease stage is reached. In these patients, as well as attention to respiratory status, vigilance is required for the possibility of the re-feeding syndrome as a post-operative complication (Fotheringham et al. 2005).

Respiratory support. Respiratory muscle weakness develops insidiously during the course of motor neurone disease, causing dyspnoea, orthopnoea, and symptoms of carbon dioxide retention, which include daytime somnolence, morning headaches, and lack of restorative sleep, with frequent waking. The management of the respiratory complications include the following general measures. Attention should be given to the detection and prevention of aspiration pneumonia. Antiobiotic therapy should be used at the first indication of a chest infection. When the patient has difficulty in clearing secretions from the chest, chest physiotherapy and postural drainage should be used if possible. The provision of a suction machine and the prescription of a mucolytic agent such as carbocisteine to reduce the viscosity of secretions may also be helpful. Patients will breathe more comfortably during sleep if placed in a semi-upright position. When patients experience bouts of severe dyspnoea, accompanied by extreme anxiety or panic, a small dose of lorazepam may be useful, 0.5–1 mg sublingually. If breathlessness causes distress during the later stages of the disease, the use of small amounts of morphine will be useful. Further depression of respiration can usually be avoided if the initial dose is small and increments are gradual.

Assisted ventilation, coupled with appropriate nutritional support, could theoretically extend the patient’s life indefinitely, and the implications of initiating such respiratory support must be clearly thought through and discussed for each patient. There are considerable international differences in the use of assisted ventilation to manage respiratory failure in amyotrophic lateral sclerosis. The progressive nature of the condition has acted as a deterrent, in some countries, for the active management of respiratory dysfunction in many patients, particularly when other motor disabilities are extensive. Full 24-h intermittent positive pressure ventilation via a tracheostomy, is an option that is chosen only rarely by fully informed patients. The costs of tracheostomy ventilation, in terms both of financial resources and the caregiver support required, are substantial. Non-invasive intermittent positive pressure ventilation via a mask (Fig. 23.5) is a practical option for respiratory support. Non-invasive positive pressure ventilation used overnight has been shown to alleviate symptoms of chronic hypoventilation and to significantly improve several measures of quality of life (Lyall et al. 2001; Bourke et al. 2003). In the United Kingdom only a small proportion of patients with amyotrophic lateral sclerosis are treated with non-invasive positive pressure ventilation, and there is marked variation in clinical practice (Bourke et al. 2002). This may partly be due to regional variation in the availability of non-invasive positive pressure ventilation. The early signs and symptoms of hypoventilation are also subtle and easily overlooked.

 Different types of mask which can be used in non-invasive ventilation, including nasal mask, a mask that avoids pressure over the bridge of the nose and a full facial mask.
Fig. 23.5

Different types of mask which can be used in non-invasive ventilation, including nasal mask, a mask that avoids pressure over the bridge of the nose and a full facial mask.

Recent work has demonstrated that the criteria most predictive of symptomatic benefit from non-invasive positive pressure ventilation in patients with motor neurone disease are: orthopnoea, daytime hypercapnia, nocturnal oxygen desaturation, together with relatively preserved bulbar function (Bourke et al. 2003). A recently published randomized, controlled trial of non-invasive ventilation in motor neurone disease has shown that in patients without severe bulbar dysfunction, survival is significantly extended with a median survival benefit of approximately 7 months, with maintenance of and improvement in multiple quality of life measures (Bourke et al. 2006). The survival benefit from non-invasive positive pressure ventilation in this group is much greater than from currently available neuroprotective therapy. Patients with severe bulbar dysfunction have more difficulty in tolerating non-invasive positive pressure ventilation for hours at a time and in this group there was some improvement in sleep-related symptoms but no demonstrable survival benefit.

End of life care. In United Kingdom practice the opening up of hospice places for patients with amyotrophic lateral sclerosis has greatly improved the quality of care and support for patients in the later stages of the disease. If motor neurone disease patients are not ventilated, they will almost always die in their sleep from hypercapnic coma. In the terminal phases of illness the aim of treatment is to ensure that the patient is comfortable, and opiate and anxiolytic medication should be used as required to alleviate discomfort or distress.

There is no therapy currently available which has a dramatic effect in slowing disease progression in amyotrophic lateral sclerosis. However, some small steps have been made towards this ultimate goal in recent years. An understanding of the molecular pathways that lead to motor neurone death (Section 23.2.4), is needed in order to target therapeutic strategies. These insights into the mechanisms of neuronal degeneration have led to the development of a number of compounds which protect neurones in cell culture and in animal models of motor neurone disease. Over 50 potential neuroprotective agents have been tested in clinical trials which have been extensively reviewed elsewhere (Meininger et al. 2000; Turner and Leigh 2003). The larger recent trials, and their theoretical and experimental basis are summarized in Table 23.6.

Table 23.6
Recent clinical trials in amyotrophic lateral sclerosis targetting different postulated disease mechanisms
Therapeutic agentRationaleResult of clinical trial in ALS patients

Excitotoxicity

Riluzole

Sodium channel blocker that inhibits presynaptic glutamate release. Slowed disease progression in SOD1 mouse model of amyotrophic.

Modest significant survival benefit

Branched chain amino acids

Activate glutamate dehydrogenase to reduce glutamate levels

No significant benefit.

Gabapentin

Reduces glutamate activity. Slowed disease progression in SOD1 mutant mouse models of motor neurone disease.

Trend towards slowing of disease progression in pilot trial not duplicated in further trial.

Topiramate

Reduces glutamate activity. Protects against motor neurone degeneration in vitro.

No significant benefit

Oxidative stress

Vitamin E (α-tocopherol)

Supplementation of the diet of SOD1 transgenic mice with vitamin E delayed onset of symptoms and slowed disease progression

No survival benefit. Significantly more patients remained in a milder disease state after 12 months of treatment.

N-acetylcysteine

N-acetylcysteine is a precursor of the antioxidant glutathione

No significant difference in survival or disease progression in an under-powered trial.

Neurotrophic factors

CNTF (subcutaneous)

These neurotrophic factors promote survival of motor neurones in vitro and arrest disease progression in the wobbler mouse model of motor neuron disease.

No benefit shown, detrimental effect at higher doses

BDNF (subcutaneous and intrathecal)

No significant benefit. Intrathecal trial terminated early due to increased incidence of adverse events in the treated group (unpublished)

IGF-1 (subcutaneous)

Promotes motor neurone survival in several models of neuronal injury

Significant slowing of disease progression in a US trial not duplicated in a European study. Cochrane review concluded IGF-1 use could not be recommended.

Mitochondrial dysfunction

Creatine

Phosphocreatine allows the rephosphorylation of ADP to ATP. Oral creatine supplementation may improve cellular energy deficits, and prolongs survival in SOD1 transgenic mice.

No significant benefit

Miscellaneous compounds

Xaliproden

Oral neurotrophic agent. Neurotrophic effects in animal models of neurodegeneration

No significant benefit

Pentoxifylline

A phosphodiesterase inhibitor already used in the treatment of peripheral vascular disease. Identified as a potential therapeutic target for amyotrophic lateral sclerosis through screening in transgenic mice.

No significant benefit

Ono-2506

An inhibitor of astrocyte activation. Neuroprotective effects in cell culture and animal models of neuronal injury.

Overall phase II trial showed no significant benefit, but post-hoc analysis indicated possible benefit in patient subgroup early in the disease course

Novartis TCH346

Prevents neuronal apoptosis

No significant benefit

Therapeutic agentRationaleResult of clinical trial in ALS patients

Excitotoxicity

Riluzole

Sodium channel blocker that inhibits presynaptic glutamate release. Slowed disease progression in SOD1 mouse model of amyotrophic.

Modest significant survival benefit

Branched chain amino acids

Activate glutamate dehydrogenase to reduce glutamate levels

No significant benefit.

Gabapentin

Reduces glutamate activity. Slowed disease progression in SOD1 mutant mouse models of motor neurone disease.

Trend towards slowing of disease progression in pilot trial not duplicated in further trial.

Topiramate

Reduces glutamate activity. Protects against motor neurone degeneration in vitro.

No significant benefit

Oxidative stress

Vitamin E (α-tocopherol)

Supplementation of the diet of SOD1 transgenic mice with vitamin E delayed onset of symptoms and slowed disease progression

No survival benefit. Significantly more patients remained in a milder disease state after 12 months of treatment.

N-acetylcysteine

N-acetylcysteine is a precursor of the antioxidant glutathione

No significant difference in survival or disease progression in an under-powered trial.

Neurotrophic factors

CNTF (subcutaneous)

These neurotrophic factors promote survival of motor neurones in vitro and arrest disease progression in the wobbler mouse model of motor neuron disease.

No benefit shown, detrimental effect at higher doses

BDNF (subcutaneous and intrathecal)

No significant benefit. Intrathecal trial terminated early due to increased incidence of adverse events in the treated group (unpublished)

IGF-1 (subcutaneous)

Promotes motor neurone survival in several models of neuronal injury

Significant slowing of disease progression in a US trial not duplicated in a European study. Cochrane review concluded IGF-1 use could not be recommended.

Mitochondrial dysfunction

Creatine

Phosphocreatine allows the rephosphorylation of ADP to ATP. Oral creatine supplementation may improve cellular energy deficits, and prolongs survival in SOD1 transgenic mice.

No significant benefit

Miscellaneous compounds

Xaliproden

Oral neurotrophic agent. Neurotrophic effects in animal models of neurodegeneration

No significant benefit

Pentoxifylline

A phosphodiesterase inhibitor already used in the treatment of peripheral vascular disease. Identified as a potential therapeutic target for amyotrophic lateral sclerosis through screening in transgenic mice.

No significant benefit

Ono-2506

An inhibitor of astrocyte activation. Neuroprotective effects in cell culture and animal models of neuronal injury.

Overall phase II trial showed no significant benefit, but post-hoc analysis indicated possible benefit in patient subgroup early in the disease course

Novartis TCH346

Prevents neuronal apoptosis

No significant benefit

Riluzole. Has been shown to significantly slow disease progression and is the only neuroprotective agent licensed for use in motor neurone disease. It is a sodium channel blocker whose primary mechanism of action is to reduce excitotoxicity through inhibition of glutamate release. Also it has been shown to have several other potentially neuroprotective effects. Two double-blind placebo-controlled trials of riluzole have been carried out in more than 1100 patients (Bensimon et al. 1994; Lacomblez et al. 1996). A Cochrane review of riluzole therapy in motor neurone disease concluded that there is a statistically significant, although modest, effect in prolonging survival by approximately 3 months (Miller et al. 2002). No clear effect on muscle strength was demonstrated, and neither trial evaluated quality of life. In view of the high cost to benefit ratio, there has been controversy about the use of riluzole worldwide. In the United Kingdom, its use is recommended by the National Institute for Clinical Excellence, which estimated the cost of therapy to be £34 000 to £43 500 per quality-adjusted life year, or QALY. Riluzole therapy is relatively expensive and the average survival benefit to be expected is modest, but against this must be weighed the arguments that the patient population requiring the drug is relatively small, and that these patients are facing a lethal disease for which no other therapy is available. At presentit is unknown whether the modest overall therapeutic effect ofriluzole conceals individual good responders and non-responders.

Other neuroprotective agents. Several compounds that appeared to protect neurones from degeneration in cell culture and animal models have had disappointing results in human clinical trials. There are two possible explanations for this. First, the models used may not accurately reproduce human disease or the testing in the models may be insufficiently rigorous. Several drugs effective in SOD1 transgenic mice have not been beneficial in human trials, including gabapentin, creatine, topiramate, and vitamin E. Potential explanations for failure of translation into effective human neuroprotective therapies include starting therapy presymptomatically in mice, and deficiencies in the design of mouse trials including failure to take into account gender and litter effects. Also problems with the design and methodology of human clinical trials in the past could mask a modest clinical benefit. To improve the design and implementation of clinical trials in motor neurone disease, the World Federation of Neurology published consensus guidelines in 1998 (Miller et al. 1999).

Future developments. The next few years are likely to see further progress in defining the molecular mechanisms of cell death underlying the neurodegenerative process in amyotrophic lateral sclerosis. It can be anticipated that further genetic mutations associated with familial disease and the contribution of genetic factors to the sporadic form of the disease will be identified. The continuing use and refinement of cellular and animal models will allow researchers to understand the sequential molecular events leading to motor neurone cell death and to evaluate new neuroprotective strategies.

Several potential neuroprotective agents are being evaluated or will shortly be entered into clinical trials in amyotrophic lateral sclerosis. These include minocycline, arimoclomol, glatiramer acetate, co-enzymeQ10, celecoxib, and ONO-2506. Automated laboratory assays of neurodegeneration can rapidly screen thousands of chemicals to identify lead compounds for drug development. The traditional reluctance of pharmaceutical companies to invest heavily in rarer diseases has recently been addressed by the emergence of non-profit-making biotech companies and academic institutions using high-throughput drug screening to identify compounds of interest. Future neuroprotective therapy for patients with amyotrophic lateral sclerosis may well involve a ‘cocktail’ of pharmacological agents aimed at different mechanisms contributing to the biochemical cascade of cell injury. Gene therapy approaches using viral vectors pseudotyped to ensure retrograde transport within motor neurone axons following intramuscular injection have shown great promise in murine models of motor neurone disease (Azzouz et al. 2004) and phase 1 human trials are expected in the near future. Cell replacement therapy using stem cells poses particular difficulties in relation to amyotrophic lateral sclerosis, not least because of the long axonal processes required for the normal function of motor neurones and the inhibitory signals preventing the effective growth and correct synaptic alignment of newly generated motor neuronal axons. Perhaps the earliest promise of cell replacement therapy for motor neurone disease will be to try to create a supportive environment of non-neuronal cells in the vicinity of motor neurones to achieve neuroprotection of existing differentiated motor neurones. Measures to improve supportive care for patients and their families will continue to develop including the judicious use of ventilatory support. As clinical and scientific developments allow the prospect of increasing the duration of the disease and prolonging survival of patients with motor neurone disease, very careful attention needs to be paid to the quality of life of afflicted individuals.

The term spinal muscular atrophy encompasses a group of genetically determined pure lower motor neurone disorders in which degeneration of the anterior horn cells leads to progressive, symmetrical muscle weakness, and wasting, with sparing of sensation, and absence of pyramidal tract involvement. As the bulbar musculature may be affected, and motor neurone degeneration is therefore not confined to the spinal cord, an alternative term ‘hereditary motor neuronopathy’ was proposed, and both terms are currently in use. There are difficulties with the definition and classification of spinal muscular atrophy, which are gradually being resolved as the underlying genetic defects are identified. By definition spinal muscular atrophy is genetically determined, but adult patients sometimes present with what appears to be a sporadic form, and a genetic basis is therefore unproven. The neurodegenerative process in spinal muscular atrophy has a predilection for motor neurones, but in some families, pyramidal tract and sensory involvement are also seen. Thus the condition shows some overlap with other neurodegenerative diseases affecting the central and peripheral motor systems.

This is the most common type of spinal muscular atrophy. It is inherited as an autosomal recessive disorder and is one of the most common lethal childhood autosomal recessive diseases. Patients develop predominantly proximal limb weakness, with relative sparing of the facial muscles and the diaphragm. Proximal spinal muscular atrophy is divided into subtypes, according to severity and age of onset (Zerres and Rudnik-Schoneborn 1995):

Type I spinal muscular atrophy, or Werdnig–Hoffman disease, presents with severe generalized muscle weakness and hypotonia at birth, or by the age of 6 months. Affected children never sit or walk, and usually die from respiratory insufficiency within the first 2 years of life.

Type II spinal muscular atrophy is an intermediate form with onset of muscle weakness before the age of 18 months, and patients can sit, but are never able to walk unaided, and survival is usually limited to adolescence.

Type III spinal muscular atrophy, or Wohlfart–Kugelberg–Welander disease, presents after the age of 18 months. Patients gain the ability to stand and walk, but often become wheelchair-dependent in adolescent or adult life though life expectancy is normal.

Onset of recessive proximal spinal muscular atrophy has been described in adulthood, and this is sometimes designated type IV spinal muscular atrophy.

Linkage analysis revealed that the three subtypes of proximal recessive spinal muscular atrophy mapped to chromosome 5q13. This region of chromosome 5 is complex, and characterized by low copy repeats, which may account for instability of this region, and trigger frequent deletions or gene conversions. Within the spinal muscular atrophy critical region, there is a 500 kB inverted duplication, with four genes present in at least two copies, telomeric and centromeric: the survival motor neuron gene, SMN, the neuronal apoptosis inhibitory protein gene, NAIP, the gene encoding BTF2p44, a subunit of RNA polymerase II involved in transcription, and a putative RNA binding protein, H4F5. Homozygous deletion of the telomeric copies of all four genes have been described in patients with spinal muscular atrophy, but the frequency of gene deletions is much higher for SMN1 than for the other 3 genes and it is now well established mutations affecting SMN1 cause 5q13-linked spinal muscular atrophy. In a series of 525 patients with classical spinal muscular atrophy, 96 per cent were linked to chromosome 5q13, and all of these showed mutations in SMN1 (Wirth et al. 2000). Furthermore, mice possess only one survival motor neuron gene, Smn, loss of which is embryonically lethal. However, Smn−/−;SMN2 mice that carry one or two copies of human SMN2 develop a phenotype of motor neuron degeneration that resembles spinal muscular atrophy in type (Monani et al. 2000).

Duplication of the SMN gene occurred more than 5 million years ago, before the separation of human and chimpanzee lineages and subsequent sequence divergence in Homo sapiens has led to 5 base pair differences between SMN1, and its centromeric homolog SMN2. The primary gene sequences of SMN1 and SMN2 predict identical proteins, but the translationally silent change in exon 7 of SMN2 decreases the activity of an exonic splicing enhancer, leading to skipping of exon 7 and a truncated protein in 80 per cent of the transcript produced by SMN2. Homozygous absence of SMN2, found in about 5 per cent of controls, has no clinical phenotype. The majority of 5q13-linked spinal muscular atrophy patients show homozygous absence of SMN1 exon 7. This may occur through gene deletion, often a large deletion that includes the whole gene, or several genes within the critical region for spinal muscular atrophy. Alternatively, SMN1 may be replaced by a copy of SMN2 during DNA replication, a process known as gene conversion.

Multiple more subtle intragenic mutations have been described in spinal muscular atrophy, the majority of which produce a truncated protein, either through splice site mutations that disrupt exon 7, or through nonsense or frameshift mutations which introduce a stop codon. Missense mutations show an interesting pattern of clustering which provides some insight into the functional domains of the SMN protein. A tyrosine–glycine rich sequence at the C-terminal of SMN encompasses five of the described missense mutations, and is a highly conserved sequence, identical in yeasts and nematodes. This region has homology to RNA interacting proteins. A further cluster of mutations is seen in the central region of SMN, which constitutes a so-called Tudor domain, an evolutionarily conserved sequence of unknown function found in many eukaryotic proteins.

SMN2 produces only 20 per cent of full-length protein. This is insufficient to rescue the phenotype in homozygous deletion of SMN1, but there is strong evidence that SMN2 is a disease-modifying gene for spinal muscular atrophy. A molecular basis for the wide variation in the severity of the phenotype resides in the fact that homozygous absence of SMN1 can be due to gene deletion or to conversion to SMN2. A patient may have anything from 1 to 4 copies of SMN2, leading to a progressive increase in the amount of full-length protein. Both the copy number of SMN2 and the protein levels of SMN have been shown to correlate with severity of disease phenotype (Feldkotter et al. 2002; Lefebvre et al. 1997). However, the copy number of SMN2 in types I, II, and III spinal muscular atrophy overlaps, therefore this alone cannot expain the phenotypic variation of the disease, and other disease-modifying factors must exist.

The majority of type I spinal muscular atrophy patients display large scale 5q13 deletions, removing SMN1 and adjacent microsatellite markers, whereas type III patients tend to have small deletions or gene conversions affecting only SMN1(Rodrigues et al. 1996). This suggests that a spinal muscular-atrophy-modifying locus distinct from SMN1 lies in the 5q13 interval. NAIP is a good candidate as it functions as a negative regulator of apoptosis. NAIP deletion occurs in 45 per cent of type I spinal muscular atrophy patients, and 18 per cent of type II and III patients, therefore loss of NAIP may lead to a more severe disease phenotype (Roy et al. 1995). H4F5 lies closer to SMN1 than any other known gene. Homozgous deletions of this gene have been found in 90 per cent of type I spinal muscular atrophy cases, and it has also been proposed as a disease-modifying gene. However, deletions of both NAIP and H4F5 may simply reflect larger chromosomal deletions that involve both SMN1 and SMN2, and their role in the pathogenesis is not confirmed.

SMN produces a protein of 294 amino acids that is widely expressed. In spinal muscular atrophy patients, the level of the SMN protein is only moderately reduced in muscle and lymphoblasts, but is reduced 100-fold in the spinal cord of type I patients (Coovert et al. 1997). SMN protein shows diffuse cytoplasmic expression, but within the nucleus, is clustered in suborganelles called ‘gems’, for ‘gemini of coiled bodies’. These are similar in size and number to, and often associated with Cajal or coiled bodies, which are known to have a role in mRNA metabolism. The SMN protein oligomerizes, and associates with six proteins named Gemins, to form the SMN complex. Self-association occurs through an oligomerization domain in exon 6, and appears to be essential for its activity (Lorson et al. 1998).

The SMN complex interacts with several proteins, many of which are involved in RNA metabolism. These are ubiquitous cellular processes, which would indicate that the clinical features of spinal muscular atrophy may be caused by a particular susceptibility of lower motor neurones to defects in RNA handling. Alternatively, SMN may have as yet unidentified functions that are specific to the motor neurone. The list of proteins reported to interact with SMN also includes several which are not involved in RNA metabolism, including profilin, the FUSE binding protein, ZRP1, and p53. The functional significance of these interactions is currently unknown.

Within the cytoplasm, the SMN complex has an important role in the assembly of spliceosomal small nuclear ribonucleoproteins, snRNPs (Pellizzoni et al. 1998; Yong et al. 2004). SMN has also been shown to have a function in pre-mRNA splicing in the nucleus. SMN mutations found in patients with spinal muscular atrophy cause deficiencies in splicing regeneration activity and interactions with other key proteins, but it is uncertain whether the function of SMN in pre-mRNA splicing explains the motor neurone specific pathology in spinal muscular atrophy. The SMN complex also interacts with viral transcriptional activators, and with RNA polymerase II, pol II, which physically and functionally couples transcription, splicing, and polyadenylation, an association mediated by RNA helicase A, RHA. Expression of a dominant-negative mutant of SMN causes accumulation of pol II and RHA in the nucleus, and inhibits transcription in vivo, suggesting a role for SMN in transcriptional regulation (Pellizzoni et al. 2001).

Motor neurone-specific functions of SMN. SMN has been shown to interact with two heteronuclear ribonucleoproteins, hnRNP-R and hnRNP-Q, which are considered to play important roles in mRNA editing, transport, and splicing. HnRNP-R is predominantly located in the axons of motor neurones, where it colocalizes with SMN, a finding which led to the confirmation of a motor-neurone-specific function of SMN (Rossoll et al. 2002). In zebrafish, knock-down of SMN protein levels in the developing embryo, caused pathfinding defects specific to the motor axon (McWhorter et al. 2003). Similarly, primary motor neurones cultured from a transgenic spinal muscular atrophy mouse model show reduced axon growth, while overexpression of SMN and hnRNP-R in cultured neuronal cells promoted neurite outgrowth (Rossoll et al. 2003). These findings raise the possibility that SMN is involved in transport of mRNA molecules in the axons of motor neurones.

Other disorders may present in infancy with hypotonia and a pattern of weakness identical to Werdnig–Hoffman disease, which are distinguished by associated features. The aetiological relationship of these disorders to classical spinal muscular atrophy has been clarified by testing for SMN mutations:

Spinal muscular atrophy with pontocerebellar hypoplasia presents with neonatal hypotonia, nystagmoid eye movements, cortical blindness, and mental retardation. Affected patients do not have mutations in SMN (Rudnik-Schoneborn et al. 2003).

Spinal muscular atrophy and arthrogryposis. Arthrogryposis, congenital joint contractures, is caused by decreased foetal movements in utero that can occur in the context of several underlying problems, including neuropathies, myopathies, and oligohydramnios. Some infants with 5q13-linked spinal muscular atrophy have arthrogryposis, the presence which was previously regarded as an exclusion criterion for spinal muscular atrophy.

Spinal muscular atrophy with arthrogryphosis and bone fractures is characterized by a pattern of weakness indistinguishable from spinal muscular atrophy type 1, and congenital long-bone fractures (Kelly et al. 1999). It is genetically distinct from SMN-related spinal muscular atrophy, and may be autosomal recessive or X-linked recessive.

X-linked spinal muscular atrophy/arthrogryphosis is linked to the short arm of chromosome X. Affected infants have congenital joint contractures, facial dysmorphia, chest deformities, hypotonia, and areflexia, and electromyographic studies and muscle biopsy consistent with loss of lower motor neurones (Kobayashi et al. 1995).

Lethal congenital contracture syndrome 1 is a disorder of multiple congenital contractures with neuropathological changes resembling those of spinal muscular atrophy, restricted to Finland. The disorder is fatal in the third trimester, and has associated features of intrauterine growth retardation, foetal hydrops, and facial abnormalities. Linkage has been established to chromosome 9p34 (Makela-Bengs et al. 1998).

Lethal congenital contracture syndrome 2. A similar syndrome, affecting Israeli Bedouins, is distinguished by additional craniofacial and ocular findings, lack of hydrops, multiple pterygia, fractures, and bladder abnormalities. Linkage to both 5q13 and 9p34 have been excluded (Landau and Mishori-Dery et al. 2003).

A missense mutation, P56S, in the vesicle-associated membrane protein, VAPB gene on chromosome 20q13.3 was identified in seven kindreds, with three different phenotypes of autosomal dominant motor neurone disease (Nishimura et al. 2004). Some patients had an atypical slowly progressive form of amyotrophic lateral sclerosis with tremor, ALS 8, some had typical rapidly progressive familial amyotrophic lateral sclerosis, while others presented with adult onset proximal spinal muscular atrophy, Finkel Type. VAPB is discussed further in Section 23.2.5.

The distal distal spinal muscular atrophies or the distal hereditary motor neuronopathies, or dHMN, are a more diverse group of disorders. They commonly manifest as a peroneal muscular atrophy syndrome, distinct from the Charcot–Marie–Tooth syndrome (Section 21.4), which causes a similar pattern of weakness, by the lack of sensory involvement. The clinical picture is one of progressive weakness of the toes and feet which may be associated with foot deformity and which extends over time to involve the distal upper limb muscles. Some patients have unusual or additional features, including predominant involvement of the hands, vocal cord paralysis, diaphragm paralysis, and pyramidal tract signs. Harding proposed seven subtypes of distal hereditary motor neuronopathy (Harding 1993; European CMT Consortium 1998), on the basis of genetic and clinical criteria. This classification was reviewed by the European Charcot–Marie–Tooth Consortium in 1998, but this is constantly evolving with the identification of novel clinical and genetic entities, the finding that previously delineated phenotypes show genetic heterogeneity, and that single gene disorders can vary widely in phenotype (Irobi et al. 2004).

In a series of 200 patients with infantile-onset spinal muscular atrophy, Rudnik-Schoneborn et al. (1996) found that approximately 1 per cent had diaphragmatic weakness and did not have deletions of the SMN gene on chromosome 5q. This subtype of infantile spinal muscular atrophy, known by the acronym SMARD, is characterized by severe breathing difficulties and limb weakness which is predominantly distal. SMARD1 is caused by mutations in the gene encoding immunoglobulin mu-binding protein 2, IGHMBP2, which has RNA helicase activity, and is involved in pre-mRNA processing (Grohmann et al. 2001). Some affected infants have evidence of sensory and autonomic nerve involvement as well as the motor deficit. Mutations in the homologous murine gene, ighmbp2 are responsible for spinal muscular atrophy in the ‘neuromuscular degeneration’, nmd mouse, which shows close phenotypic resemblance to SMARD1 (Cox et al. 1998).

This disorder causes slowly progressive distal limb weakness and has a very variable age of onset. In the most severely affected patients, there is evidence of diaphragmatic involvement, with a decrease in vital capacity and elevation of the hemidiaphragms on chest radiographs. In a large consanguineous Lebanese pedigree, genetic linkage has been mapped to chromosome 11q though mutations in IGHMBP2 have been excluded. A distinct form of autosomal recessive distal spinal muscular atrophy, identified in families from the Jerash region of Jordan, has been mapped to chromosome 9p21.1-p12.

Pedigrees with distal spinal muscular atrophy with upper limb predominance, designated dHMN-V, often include affected individuals with evidence of mild sensory disturbance or upper motor neurone features. The disorder was linked to chromosome 7p in a large Bulgarian family (Christodoulou et al. 1995). Subsequently a Mongolian family was identified in which Charcot–Marie–Tooth disease type 2D and distal spinal muscular atrophy with upper limb predominance segregated in the same kindred. All affected members had weakness and wasting of the intrinsic muscles of the hands. Those with no sensory deficit and peroneal muscle weakness had a diagnosis of distal spinal muscular atrophy, while in other affected individuals Charcot–Marie–Tooth disease was diagnosed on the basis of a glove and stocking sensory loss. Both disorders in this family were linked to the same region of chromosome 7p. Mutations were subsequently uncovered in the gene encoding glycyl tRNA synthetase, GARS, in Charcot–Marie–Tooth disease type 2D families, in families with dHMN-V, and in the family described with both disorders (Antonellis et al. 2003). Identification of the underlying genetic disorder has therefore confirmed that Charcot–Marie–Tooth Type 2D and distal spinal muscular atrophy with upper limb predominance are phenotypic variants of a single disorder. GARS is a member of the family of aminoacyl tRNA sythetases involved in diverse cellular processes, including charging tRNAs with their appropriate amino acids. This defect would be expected to affect every glycine-containing protein, and it is at present unknown why this gene defect specifically impairs the function of neurones.

Linkage to chromosome 7p15 was excluded in a large Austrian family with dHMN-V. In this family and several others, heterozygous mutations were identified in the Berardinelli–Seip congenital lipodystrophy gene, (Windpassinger et al. 2004). The gene encodes seipin, an integral membrane protein of the endoplasmic reticulum, and null mutations cause Berardinelli–Seip congenital lipodystrophy. N88S and S90L mutations affect glycosylation of seipin, resulting in the formation of intracellular aggregates. The same mutations were also found to be associated with the Silver syndrome variant of hereditary spastic paraplegia (Section 23.4.2). This is a further example of the same genetic defect causing two quite distinctive phenotypes. The clinical features associated with BSCL2 mutations have recently been broadened to include individuals who have lower limb predominant distal amyotrophy in the absence of pyramidal tract signs, and some with a combination of spasticity and severe amyotrophy in the lower limbs (Irobi et al. 2004).

Designated dHMN-II, this is usually inherited as an autosomal dominant trait, but sporadic cases are frequently described, which may reflect late-onset recessive disease, non-genetic aetiology, or new mutations. One large Belgian pedigree was linked to chromosome 12q24 and subsequently heterozygous mutations were found in the gene encoding the small heat shock 22 kDa protein 8, HSPB8/HSP22, in four families (Irobi et al. 2004c). Missense mutations have also been identified in the interacting partner of HSP22, heat shock 27kDa protein 1, HSPB1/HSP27, both in patients with dHMN-II and in pedigrees with Charcot–Marie–Tooth Type 2F (Evgrafov et al. 2004). Most of the HSP22 and HSP27 mutations disrupt a conserved αcrystallin domain in these proteins and cell biological studies have shown that HSP22 mutants show greater binding to HSP27, resulting in the formation of intracellular aggregates (Irobi et al. 2004a). HSP27 is also involved in the organization of the neurofilament network, important for the maintenance of the axonal cytoskeleton and for axonal transport, and mutant HSP27 perturbs neurofilament assembly (Evgrafov et al. 2004).

Distal spinal muscular atrophy with vocal cord paralysis, designated dHMN-VII, characterized by distal limb weakness and dysphonia, has been described to belinked to chromosome 2q14 in two large Welsh families with common ancestry (McEntagart et al. 2001). Three disorders have been described which show similar clinical features to dHMN-VII: one family with distal spinal muscular atrophy and vocal cord paralysis also had sensorineural hearing loss, (Bolthauser et al. 1989) while Charcot–Marie–Tooth disease type IIC is characterized by progressive vocal cord paralysis, distal limb weakness, and sensory loss (Donaghy and Kennett 1999). The gene alterations underlying these two disorders are currently unknown. A third disorder, presenting with breathing difficulty due to vocal cord paralysis, progressive facial weakness, and weakness and atrophy of the hands, has been found to be caused by a missense mutation in the gene encoding dynactin, DCTN1, on chromosome 2p13. This mutation is predicted to distort the folding of the dynactin microtubule binding domain, and lead to dysfunction of dynactin-mediated retrograde axonal transport (Puls et al. 2003).

This relatively benign disorder, described in pedigrees from Holland and Canada, has been shown by linkage analysis to map to chromosome 12q23-24 (van der Vleuten et al. 1998). Again, the disorder is genetically heterogeneous, as in one family linkage to this region has been excluded.

Muscle weakness and wasting in a scapuloperoneal distribution may be myopathic or neurogenic in origin. Several large pedigrees with neurophysiological evidence of denervation have been described. In one pedigree, linkage has been mapped to chromosome 12q24.1-12q24.31 (Isozumi et al. 1996) which is close to the region of chromosome 12 to which a myopathic scapuloperoneal syndrome is linked (Wilhelmsen et al. 1996). Interestingly many of the families described with scapuloperoneal spinal muscular atrophy include individuals with both myopathic and neurogenic changes in affected muscles. Congenital non-progressive spinal muscular atrophy affecting the lower limbs maps to the same region as scapuloperoneal spinal muscular atrophy, but it has not yet been established whether these disorders are allelic.

Monomelic amyotrophy has been chiefly reported from Japan and India. Hiroyama et al. (1959) reported unilateral atrophy of the upper limb and gave it the term ‘juvenile muscular atrophy of unilateral upper extremity’. Later a report from India described cases of atrophy of the muscles of one lower limb and described it as the ‘wasted leg syndrome’ (Prabhakar et al. 1981). Gourie-Devi et al. (1984) suggested the term monomelic amyotrophy to cover both of these entities. Monomelic amyotrophy has been reported to account for 8–29 per cent of all motor neuron diseases in the series reported from India (Gourie-Devi et al. 1984; Saha et al. 1997).

Monomelic amyotrophy is a benign variant of motor neurone disease that predominantly affects young men. Lower motor neurone features of wasting and weakness are usually confined to one upper or less commonly the lower limb, without involvement of other components of the nervous system. Patients with the upper limb variant often report tremulousness of the fingers. In the upper limb, the muscles innervated by the C7 to T1 spinal segments i.e. the intrinsic hand muscles and flexors and extensors of the wrists and fingers, tend to be most severely affected. Relative sparing of the brachioradialis muscle amongst the surrounding atrophic forearm muscles is a characteristic feature of this condition (Hirayama et al. 1963). The disorder is usually sporadic, although rarely it may be familial (Nalini et al. 2004). In the lower limb variant, the muscle atrophy most commonly involves both proximal and distal muscles, but some patients have predominant involvement of either the thigh or lower leg musculature (Prabhakar et al. 1981; Gourie-Devi et al. 1984). Muscle cramps and fasciculations are observed in 20–30 per cent of patients and unilateral pes cavus may be a presenting feature in the lower limb variant.

Some patients develop similar symptoms in the contralateral limb, but the disorder usually remains strikingly asymmetrical. In most cases, the onset is insidious, with a slow progression over 2–4 years, followed by a plateau phase, although further weakness may evolve for up to 8 years (Peiris et al. 1989; Gourie-Devi and Nalini 2003a). The tendon reflexes are depressed. Seldom, if ever, does weakness spread to involve other parts of the body, and upper motor neurone signs are absent. It does not evolve to amyotrophic lateral sclerosis.

A neurogenic pattern on electromyography, and histological evidence of neurogenic atrophy of muscle indicate anterior horn cell pathology. Pathological studies in two patients, who died of coincidental causes, showed atrophy of the affected region of the spinal cord with severe, asymmetrical, bilateral loss of anterior horn motor neurones (Hirayama et al. 1987; Araki et al. 1989). Imaging studies have shown evidence of forward displacement of the dural sac during neck flexion in patients with Hirayama syndrome, and this has led to the proposal that the condition may be caused by intermittent compression and ischaemia of the cervical spinal cord. However, other reports have failed to confirm this (Schroder et al. 1999; Willeit et al. 2001) and thus the pathogenic mechanisms underlying Hirayama syndrome and other forms of monomelic amyotrophy remain uncertain. Mutations in the SMN and SOD1 genes have been excluded. A mitochondrial DNA mutation has been reported from Italy in one patient with monomelic amyotrophy and sensorineural hearing loss (Fetoni et al. 2004).

The GM2 gangliosidoses are a group of recessively inherited disorders in which deficiency of the lysosomal enzyme, β hexosaminidase A, leads to abnormal intracellular accumulation of lipids in neurons and glia (Section 10.4.2). Tay-Sachs disease, the classical infantile form of GM2 gangliosidosis associated with mutations in both alleles of the HEXA gene, is the most frequent form of the disorder and is particularly found amongst Ashkenazi Jews. Affected children, after a few months of normal development, start to regress, lose head control and become hypotonic and apathetic, with the development of seizures and cortical blindness. Examination will frequently reveal a large head, a macular cherry red spot, spasticity, quadriparesis, and an exaggerated startle response. The disorder is usually fatal before the age of 5–6 years. GM2 gangliosidosis can also occur as a later onset disease, arbitrarily divided into juvenile, early adult, and late adult forms. The late-onset forms can cause a disorder of the upper and lower motor neurons. However, there is usually evidence clinically of a multisystem disorder with involvement of the cerebellum and its connections, the autonomic nervous system, and extrapyramidal manifestatons such as Parkinsonism, dystonia, or choreoathetosis. Patients may also have evidence of peripheral neuropathy and develop features of dementia or psychosis. Pathologically the most characteristic change is the presence of swollen ballooned neurons with storage material in lysosomes and characteristic membranous cytoplasmic inclusion bodies. Meganeurites are formed which are associated with aberrant dendritic, neuritic, and synaptic growth.

Several reports have described the phenotype of motor neurone disease in adult hexosaminidase deficiency (Kaback et al. 1978; Johnson et al. 1982; Mitsumoto et al. 1985). Gudesblatt and colleagues described 52 patients who had atypical amyotrophic lateral sclerosis and 4 of these had partial hexosaminidase A deficiency (Gudesblatt et al. 1988). None of 50 patients with typical amyotrophic lateral sclerosis had abnormal HexA activity and similar findings were reported by Drory and colleagues (Drory et al. 2003). Most cases with a motor system disorder in the context of HexA deficiency have additional neurological manifestations and a pure amyotrophic lateral sclerosis phenotype is extremely rare (Karni et al. 1988; Parboosingh et al. 1997). Muscle weakness tends to develop insidiously during the second decade of life. Weakness is usually first apparent in the proximal muscles and may be associated with muscle fasciculation. Atrophy of the intrinsic hand muscles can also be seen as an early feature. Upper motor neurone features may be present or absent. As described above, the patients on close examination or on follow up over time will often have features indicative of a progressive multisystem neurodegenerative disorder.

Multifocal motor neuropathy (Section 21.11.3) is an acquired disorder which is important to distinguish from amyotrophic lateral sclerosis because it has a much more benign prognosis and is potentially amenable to therapy with intravenous immunoglobulin (Slee et al. 2007). It is characterized by slowly progressive, asymmetrical weakness which develops gradually or with stepwise progression over several years. Initially weakness may not be accompanied by significant muscle wasting. The age of onset is usually between 20 and 50 years and the disorder is more common in men. Weakness is more common in the upper limbs compared to the legs and is usually distal. Common initial symptoms include wrist drop and weakness of grip. Weakness is often more pronounced than would be expected for the degree of muscle wasting which is a clinical clue to the presence of conduction block. However, atrophy may become pronounced in patients with long duration disease. Muscle cramps and fasciculations are reported by two-thirds of patients. Multifocal motor neuropathy is commonly initially misdiagnosed as amyotrophic lateral sclerosis (Traynor et al. 2000). The slowly progressive disease course, the absence of upper motor neurone signs, and the presence of conduction block on neurophysiological examination, which may only be found after repeated testing, are important clinical clues pointing to the correct diagnosis. Raised titres of anti-GM1 ganglioside antibodies in serum may also be a diagnostic clue, though unfortunately the presence of these antibodies is not specific for multifocal motor neuropathy (Taylor et al. 1996).

Acute poliomyelitis is described in detail in Chapter 42.4.2. Some patients who recover partially or fully from acute poliomyelitic weakness develop a new syndrome of progressive motor deficit many years after the original illness. This post-polio syndrome may affect one-third of patients who have had acute paralytic poliomyelitis. The newly reported fatigue, pain, cramps, wasting, weakness, and functional deterioration develop in areas overtly or subclinically affected during the acute attack (Trojan and Cashman 2005). Typically, several decades elapse between the acute paralysis and the onset of post-polio syndrome, with the peak incidence 20–25 years after the original illness. Often no objective change in muscle strength can be detected on manual muscle testing over several years in patients who nevertheless complain of progressive weakness or of progressive difficulty carrying out activities of daily living. Post-polio syndrome must be distinguished from the non-specific symptoms of joint instability; nerve, root, or plexus compression; and increasing scoliosis which may be late secondary effects resulting from the original weakness. A recent systematic review of the relevant literature concluded that at present conclusions cannot be drawn regarding the functional course or prognostic factors in late-onset polio sequelae and that further work, including long-term follow-up studies of unselected patient populations are needed (Stolwijk-Swuste et al. 2005). The cause has not been completely elucidated, but it is likely to be due to distal axonal degeneration of enlarged post- poliomyelitis motor units. Care must be taken to identify coincidental neurological disorders and orthopaedic complications of longstanding weakness, limb dysfunction, and spinal deformity. Although currently there is no specific disease-modifying treatment for post-polio syndrome, an interdisciplinary management programme can be useful in controlling symptoms. There is some evidence that supervised aerobic muscle training and introduction of non-invasive ventilation for patients with respiratory impairment may be helpful measures (Farbu et al. 2006). A recent randomized controlled trial of intravenous immunoglobulin showed slight improvement in some parameters including muscle strength and SF-36 subscale vitality score, but no significant change in overall quality of life or pain (Gonzalez et al. 2006).

Primary lateral sclerosis is considered as part of the spectrum of amyotrophic lateral sclerosis (Section 23.2.1).

Hereditary spastic paraplegia, first described in the 1880s, is a group of hereditary neurodegenerative or neurodevelopmental diseases which affects approximately 1 in 10 000 individuals. The main feature of the clinical phenotype is progressive lower limb spasticity due to degeneration of the corticospinal tracts within the spinal cord. Hereditary spastic paraplegia is most commonly inherited as an autosomal dominant trait, but autosomal recessive and X-linked recessive forms also exist. The hereditary spastic paraplegia phenotype may be ‘pure’ in which the spastic paraparesis occurs in isolation or ‘complicated’ where the spastic paraparesis is one component of a much more complex neurological and/or systemic disorder. Hereditary spastic paraplegia shows extreme genetic heterogeneity. To date more than 30 genetic loci have been identified and genes have been identified at 14 of these (Table 23.7). The recent discovery of multiple genes is rapidly shaping new concepts of the cellular mechanisms of degeneration of the long axons of the corticospinal tract in hereditary spastic paraplegia. It is apparent that motor neurons provide an extreme example of the potential difficulties for a cell in trafficking, transport, and energy metabolism, and that the longest axons of the central nervous system may be specifically vulnerable to several distinct biochemical perturbations.

Table 23.7
Genetic classification of hereditary spastic paraplegia
Genome database DesignationChromosomeInheritancePhenotypeGenetic Defect

SPG1

Xq28

X-linked

Complicated

L1CAM

SPG2

Xq22

X-linked

Both

PLP

SPG3

14q11.2

AD

Pure

Atlastin

SPG4

2p22

AD

Both

Spastin

SPG5

8p12-q13

AR

Pure

SPG6

15q11.1

AD

Pure

NIPA1

SPG7

16q24.3

AR

Both

Paraplegin

SPG8

8q24

AD

Pure

KIAA0196

SPG9

10q23.3-24.2

AD

Complicated

SPG10

12q13

AD

Pure

KIF5A

SPG11

15q13-15

AR

Both

SPG12

19q13

AD

Pure

SPG13

2q24-q34

AD

Pure

HSP60

SPG14

3q27-q28

AR

Complicated

SPG15

14q

AR

Complicated

SPG16

Xq11.2

X-linked

Pure

SPG17

11q12-q14

AD

Complicated

Seipin

SPG18

Pending

SPG19

9q33-q34

AD

Pure

SPG20

13q12.3

AR

Complicated

Spartin

SPG21

15q22.31

AR

Complicated

Maspardin

SPG22

Pending

SPG23

1q24-q32

AR

Complicated

SPG24

13q14

AR

Complicated

SPG25

6q23.3-q24.1

AR

Complicated

SPG26

12p11.1-12q14

AR

SPG27

10q22.1-q24.1

AR

SPG28

14q21.3-Q22.3

AR

Pure/distal

sensory loss

SPG29

1p21.1-1p27.1

AR

Complicated

SPG30

2q37.3

AR

Pure

SPG31

2p12

AD

Pure

REEP1

SPG32

SPG33

10q24.2

AD

Pure

ZFYVE27

Genome database DesignationChromosomeInheritancePhenotypeGenetic Defect

SPG1

Xq28

X-linked

Complicated

L1CAM

SPG2

Xq22

X-linked

Both

PLP

SPG3

14q11.2

AD

Pure

Atlastin

SPG4

2p22

AD

Both

Spastin

SPG5

8p12-q13

AR

Pure

SPG6

15q11.1

AD

Pure

NIPA1

SPG7

16q24.3

AR

Both

Paraplegin

SPG8

8q24

AD

Pure

KIAA0196

SPG9

10q23.3-24.2

AD

Complicated

SPG10

12q13

AD

Pure

KIF5A

SPG11

15q13-15

AR

Both

SPG12

19q13

AD

Pure

SPG13

2q24-q34

AD

Pure

HSP60

SPG14

3q27-q28

AR

Complicated

SPG15

14q

AR

Complicated

SPG16

Xq11.2

X-linked

Pure

SPG17

11q12-q14

AD

Complicated

Seipin

SPG18

Pending

SPG19

9q33-q34

AD

Pure

SPG20

13q12.3

AR

Complicated

Spartin

SPG21

15q22.31

AR

Complicated

Maspardin

SPG22

Pending

SPG23

1q24-q32

AR

Complicated

SPG24

13q14

AR

Complicated

SPG25

6q23.3-q24.1

AR

Complicated

SPG26

12p11.1-12q14

AR

SPG27

10q22.1-q24.1

AR

SPG28

14q21.3-Q22.3

AR

Pure/distal

sensory loss

SPG29

1p21.1-1p27.1

AR

Complicated

SPG30

2q37.3

AR

Pure

SPG31

2p12

AD

Pure

REEP1

SPG32

SPG33

10q24.2

AD

Pure

ZFYVE27

Key: AD = autosomal dominant

AR = autosomal recessive

Pure hereditary spastic paraplegia shows progressive spastic paraparesis as the major feature, but other clinical features may be observed including bladder disturbance, mild distal muscle wasting, pes cavus, dorsal column dysfunction, and loss of ankle reflexes. The most common presenting symptom in pure forms is gait disturbance, the patient often complaining of lower limb stiffness, balance difficulties, or of a tendency to fall. In young children, delayed motor milestones or a tendency to walk on the toes, may be the first indications of a problem. The major features apparent on neurological examination consist of lower limb spasticity, hyperreflexia, and extensor plantar responses which may be accompanied by a mild pyramidal distribution weakness. A characteristic feature of hereditary spastic paraplegia is that the patient’s disability usually arises from prominent spasticity and any accompanying muscle weakness is often very mild.

There is considerable variation in age at onset and severity of the spastic paraparesis even within affected members of the same pedigree, suggesting that other genetic or environmental factors may impact on the phenotype observed. The reported age at onset of pure hereditary spastic paraplegia ranges from infancy to the eighth decade (Harding 1981). Several studies have indicated that an early age of onset, <35 years, tends to be associated with relatively slow disease progression, the majority of individuals retaining the ability to walk even in elderly life. In contrast late onset, >35 years, tends to show more rapid disease progression, and many patients become non-ambulant in the seventh and eighth decades of life (Harding 1981). Hereditary spastic paraplegia does not, in general, reduce life expectancy. Approximately 25–30 per cent of patients have a subclinical phenotype in which symptoms which are so mild that the condition may not be revealed without a neurological examination.

Complicated hereditary spastic paraplegia shows the spastic paraparesis as only one component of a much more complex disorder with additional clinical features (Table 23.8). Some of the complicated hereditary spastic paraplegia phenotypes are extremely rare, having been described only in single families. Certain complicated phenotypes show characteristic clinical features associated with particular hereditary spastic paraplegia loci. For example, hereditary spastic paraplegia associated with cognitive impairment is most commonly described in patients with spastin, SPG4, mutations (White et al. 2000; Webb et al. 1998). In patients with the SPG9 subtype of hereditary spastic paraplegia there is a very distinctive clinical phenotype with the development of cataracts, severe gastroesophageal reflux, and an axonal neuropathy superimposed on the spastic paraparesis (Seri et al. 1999). Pigmentary macular degeneration is an additional feature observed in patients with hereditary spastic paraplegia linked to the SPG15 locus (Hughes et al. 2001). Peripheral neuropathy has been described in patients with hereditary spastic paraplegia linked to the SPG11 and SPG14 loci (Vazza et al. 2000; Mostacciuolo et al. 2000). In the Silver variant of hereditary spastic paraplegia, SPG17, wasting and weakness of the hands, is a striking feature (Patel et al. 2001). Patients with Troyer syndrome, SPG20, have dysarthria, distal amyotrophy, short stature, and developmental delay in addition to spastic paraparesis (Patel et al. 2002).

Table 23.8
Additional clinical features in complicated hereditary spastic paraplegia (HSP)
Clinical featureHSP subtypeAdditional comments

Amyotrophy

Peroneal muscular atrophy

Amyotrophy associated with an axonal sensory and motor neuropathy (AD)

Silver syndrome

Severe wasting of the small muscles of the hand with sparing of the lower limb musculature. Linked to SPG17 (AD)

Troyer syndrome

Distal wasting in the limbs with delayed development, spastic quadraparesis, pseudobulbar palsy, choreathetosis, and short stature. Linked to SPG20 (AR).

Charlevoix–Saguenay syndrome

Similar to Troyer syndrome with additional ataxia, described in Quebec (AR)

Resembling juvenile FALS

Childhood onset (AR)

Cardiac defects

-

Associated with mental retardation

Cerebellar signs

-

Dysarthria with a mild upper limb ataxia

Deafness

Sensori-neural

X-linked

Dementia

Subcortical or cortical pattern

Dementia can occur in isolation with HSP, when it tends to be of the subcortical type, or be part of a much more complex phenotype (AR and AD). Linkage to SPG4 locus in a number of families

Endocrine dysfunction

Kallmann’s syndrome

Hypogonadotrophic hypogonadism and anosmia

Epilepsy

-

Various epileptic seizure types have been descibed incuding; absence, simple/complex partial, atonic, grand mal, and myoconic

Extrapyramidal Signs

Choreoathetosis

Dystonia and rigidity

 

Mast syndrome

Dementia, dysarthria, and athetosis in Amish people with onset in 2nd decade (AR)

Hyperekplexia

-

Neonatal hypertonia and an exaggerated startle response (AD).

Icthyosis

Sjögren–Larsson syndrome

Also with mental retardation and occasionally a pigmentary macular degeneration (AR).

Retinal changes

Optic atrophy

Retinal degeneration

Pigmentation seen in SPG15.

Kjellin syndrome

Dysarthria, upper limb ataxia, dementia, retinal degeneration +/− amyotrophy (AR).

Sensory neuropathy

Asymptomatic

Sensory neuropathy detected only on clinical examination.

Childhood onset

With painless ulcers and deformities secondary to neuropathic bone resorption.

Adult onset

Trophic skin changes and foot ulcers.

Others

SPG 1

Mental retardation, aphasia, a shuffling gait, and adducted thumbs. Caused by mutations in L1CAM gene (X-linked).

SPG 9

Bilateral cataracts, gastroesophageal reflux, and amyotrophy

Clinical featureHSP subtypeAdditional comments

Amyotrophy

Peroneal muscular atrophy

Amyotrophy associated with an axonal sensory and motor neuropathy (AD)

Silver syndrome

Severe wasting of the small muscles of the hand with sparing of the lower limb musculature. Linked to SPG17 (AD)

Troyer syndrome

Distal wasting in the limbs with delayed development, spastic quadraparesis, pseudobulbar palsy, choreathetosis, and short stature. Linked to SPG20 (AR).

Charlevoix–Saguenay syndrome

Similar to Troyer syndrome with additional ataxia, described in Quebec (AR)

Resembling juvenile FALS

Childhood onset (AR)

Cardiac defects

-

Associated with mental retardation

Cerebellar signs

-

Dysarthria with a mild upper limb ataxia

Deafness

Sensori-neural

X-linked

Dementia

Subcortical or cortical pattern

Dementia can occur in isolation with HSP, when it tends to be of the subcortical type, or be part of a much more complex phenotype (AR and AD). Linkage to SPG4 locus in a number of families

Endocrine dysfunction

Kallmann’s syndrome

Hypogonadotrophic hypogonadism and anosmia

Epilepsy

-

Various epileptic seizure types have been descibed incuding; absence, simple/complex partial, atonic, grand mal, and myoconic

Extrapyramidal Signs

Choreoathetosis

Dystonia and rigidity

 

Mast syndrome

Dementia, dysarthria, and athetosis in Amish people with onset in 2nd decade (AR)

Hyperekplexia

-

Neonatal hypertonia and an exaggerated startle response (AD).

Icthyosis

Sjögren–Larsson syndrome

Also with mental retardation and occasionally a pigmentary macular degeneration (AR).

Retinal changes

Optic atrophy

Retinal degeneration

Pigmentation seen in SPG15.

Kjellin syndrome

Dysarthria, upper limb ataxia, dementia, retinal degeneration +/− amyotrophy (AR).

Sensory neuropathy

Asymptomatic

Sensory neuropathy detected only on clinical examination.

Childhood onset

With painless ulcers and deformities secondary to neuropathic bone resorption.

Adult onset

Trophic skin changes and foot ulcers.

Others

SPG 1

Mental retardation, aphasia, a shuffling gait, and adducted thumbs. Caused by mutations in L1CAM gene (X-linked).

SPG 9

Bilateral cataracts, gastroesophageal reflux, and amyotrophy

Key: AD = autosomal dominant, AR = autosomal recessive, FALS = familial amyotrophic lateral sclerosis.

Epilepsy has been described as a feature complicating hereditary spastic paraplegia, including in families with spastin mutations (Gigli et al. 1993; Yih et al. 1993). There does not appear to be an association with particular types of seizures and the epilepsy may occur before or after the onset of the spastic paraparesis.

Dementia and cognitive impairment have been reported in complicated hereditary spastic paraplegia pedigrees (White et al. 2000) and have also been described as an isolated accompaniment to spastic paraparesis in both autosomal dominant and recessive families (Cross and McKusick 1967; Webb et al. 1998; Pridmore et al. 1995). The cognitive abnormalities observed, for example impairments of attention, perceptual speed, visuomotor coordination, and forgetfulness, are in keeping with a sub-cortical type of dementia and features suggesting major cortical involvement, such as dysphasia, agnosia, and dyscalculia, are usually absent. Several families with dementia complicating HSP have been described with spastin mutations, SPG4, (Webb et al. 1998; White et al. 2000). Affected individuals in SPG4 families may have subclinical cognitive impairment detectable by neuropsychological evaluation (Byrne et al. 2000).

Distal amyotrophy is one of the commonest additional features seen in patients with hereditary spastic paraplegia. Several pathologies can give rise to this amyotrophy including lower motor neurone loss, axonal neuropathy, and central axonopathy. Several characteristic syndromes of hereditary spastic paraplegia with amyotrophy have been described:

The commonest type with amyotrophy is also known as peroneal muscular atrophy with pyramidal features or hereditary motor and sensory neuropathy type V (Harding and Thomas 1984). It is usually transmitted as an autosomal dominant trait and develops in the second decade of life or later. Affected individuals have amyotrophy associated with axonal motor and sensory neuropathy as well as features of spastic paraparesis.

Silver syndrome consists of autosomal dominant hereditary spastic paraplegia complicated by striking amyotrophy of the hands (Silver 1966). There is variation in age at onset, ranging from childhood to late adult, and in severity. Some Silver syndrome pedigrees are linked to the SPG17 locus, but there is clearly genetic heterogeneity as other families are not linked to this site.

Troyer syndrome is an autosomal recessive form of hereditary spastic paraplegia complicated with amyotrophy of the hands and feet, pseudobulbar palsy, choreoathetosis, short stature, and mental retardation (Cross and McKusick 1967b; Auer-Grumbach et al. 1999). It was originally described as a childhood onset disorder in an Old Order Amish population in the USA. More recently, it has been suggested that Troyer syndrome should be broadened to include families with later onset, lack of movement disorder, atrophy or partial agenesis of the corpus callosum, and non-Amish origin. Troyer syndrome is linked to SPG20 and the gene encodes a protein named spartin (Patel et al. 2002).

Charlevoix–Saguenay syndrome, described in families from Quebec, appears similar to Troyer syndrome, but with the additional clinical feature of ataxia (Bouchard et al. 1978).

A further rare phenotype of recessive hereditary spastic paraplegia complicated by amyotrophy is described resembling juvenile onset familial amyotrophic lateral sclerosis. In these pedigrees a childhood onset spastic paraparesis is observed with prominent wasting of the distal musculature. Neurophysiological examination shows electromyographic changes in keeping with lower motor neurone degeneration (Bruyn et al. 1993).

Sensory neuropathy of variable severity with onset in childhood or adult life is also a common additional feature in complicated hereditary spastic paraplegia. Severely affected patients may develop chronic painless cutaneous ulcers and neuropathic bone resorption occurring in early life, but in some patients the sensory neuropathy may be subclinical and only detected with neurophysiological testing (Schady and Smith 1994).

Detailed neuropathological findings have been reported in relatively few cases of hereditary spastic paraplegia. Therefore the extent to which the known clinical and genetic heterogeneity is reflected in pathological heterogeneity remains to be defined. In addition, because genetic characterization has only recently emerged, there are few pathological reports on genetically characterized cases. The core neuropathological features of hereditary spastic paraplegia were first described by Strümpell and confirmed in a series of subsequent reports (Sack et al. 1978; Strumpell 1886). The spinal cord shows pallor of the lateral and frequently also the anterior corticospinal tracts, with loss of axons and myelin which most markedly affects the longest descending axons in the lumbosacral region (Fig. 23.6). There is also pallor of the dorsal columns, particularly the medial fibres within the fasciculus gracilis. Involvement of the spinocerebellar tracts is described in approximately 50 per cent of cases. Depletion of Betz cells from the motor cortex is reported in some cases, but anterior horn cells in the spinal cord have usually been reported as appearing normal. Degeneration has occasionally been described in neurones of the dorsal nucleus of Clarke. Characteristically there is more severe involvement of the distal part of the corticospinal tracts and dorsal columns so that the most severe changes in these fibre pathways are observed in the lumbar and cervical cord respectively. This has led to the hypothesis that in hereditary spastic paraplegia the neurodegeneration occurs as a ‘dying back’ axonopathy, affecting the most distal part of these long axons first.

 Spinal cord pathology in hereditary spastic paraplegia showing myelin pallor of the lateral corticospinal tracts (arrow) and the medial part of the posterior columns (arrow).
Fig. 23.6

Spinal cord pathology in hereditary spastic paraplegia showing myelin pallor of the lateral corticospinal tracts (arrow) and the medial part of the posterior columns (arrow).

A recent study described the distribution of spastin in the normal human central nervous system, as well as the molecular pathology of three cases of hereditary spastic paraplegia with defined mutations in spastin (Wharton et al. 2003). Spastin was shown to be a neuronal protein widely distributed within the central nervous system. Within motor neurones, spastin was predominantly expressed in the cytoplasm of the cell body, with some extension of staining into the proximal neurites and axons. Interestingly, although not prominently affected clinically in SPG4, the spinal cord lower motor neurones showed evidence of cytopathology, with the presence of hyaline inclusion bodies some of which stained for β-tubulin; and variable loss of expression of n-phosphorylated neurofilament protein, β-tubulin, spastin, and mitochondria from the cell bodies, suggesting altered partitioning of cytoskeletal components and organelles. In addition all three cases showed evidence of tau pathology, with neurofibrillary tangles, neuropil threads, and glial tau pathology.

In many neuropathological reports of pure hereditary spastic paraplegia cerebral structures, with the exception of Betz cells, are said to be uninvolved. However subclinical involvement of other parts of the central nervous system is increasingly recognized and in particular there is emerging recognition of cognitive impairment is some families with hereditary spastic paraplegia. The neuropathological substrate of such cognitive impairment currently remains poorly defined, though isolated case reports have been described (Ferrer et al. 1995; White et al. 2000). In autosomal recessive forms of hereditary spastic paraplegia associated with mutation in the paraplegin gene, SPG7, muscle biopsies have shown ragged red fibres (Casari et al. 1998). Scattered muscle fibres show negative histochemical reaction for cytochrome oxidase, but preserved or elevated succinate dehydrogenase activity and peripheral accumulation of mitochondria. These changes, which are typical of oxidative phosphorylation defects in muscle, support a role for mitochondrial dysfunction in the pathogenesis of paraplegin mutation-associated hereditary spastic paraplegia. Skeletal muscle has not been widely surveyed in other types of hereditary spastic paraplegia, though it has been reported that muscle biopsies from some hereditary spastic paraplegia patients in whom spastin and paraplegin mutations had been excluded showed biochemical evidence of impairment in mitochondrial respiratory chain function (McDermott et al. 2003b).

Molecular genetic testing is increasingly used in the diagnosis. Although 14 causative genes have now been identified, testing for mutations in these genes is not necessary routinely available to the clinicians involved in patient care. Therefore, at present hereditary spastic paraplegia remains a diagnosis of exclusion and conditions which should be considered in the differential diagnosis are included in Table 23.9. It is clearly important to exclude diagnoses in which there is a treatable cause for the spastic paraparesis including structural lesions of the spinal cord, vitamin B12 deficiency, multiple sclerosis, or dopa responsive dystonia. It is also important to exclude other motor system disorders such as familial amyotrophic lateral sclerosis, in which the clinical course and prognosis are significantly different from those to be expected in patients with hereditary spastic paraplegia.

Table 23.9
Differential diagnosis of hereditary spastic paraplegia

Multiple sclerosis

Cerebral palsy

Spondylotic spinal disease

Amyotrophic lateral sclerosis/motor neurone disease

Other structural spinal cord disorders e.g. tumours, arteriovenous malformation

Arnold–Chiari malformation

Adrenoleukodystrophy/ adrenomyeloneuropathy

Leukodystrophies e.g. Krabbe disease, metochromatic leukodystrophy

Dopa responsive dystonia

Vitamin B12 deficiency / subacute combined degeneration of the cord

Abetalipoproteinaemia

Spinocereballar ataxias

HTLV-1 infection, tropical spastic paraparesis

Neurosyphilis

Neurolathyrism

Vitamin E deficiency

Arginase deficiency

Multiple sclerosis

Cerebral palsy

Spondylotic spinal disease

Amyotrophic lateral sclerosis/motor neurone disease

Other structural spinal cord disorders e.g. tumours, arteriovenous malformation

Arnold–Chiari malformation

Adrenoleukodystrophy/ adrenomyeloneuropathy

Leukodystrophies e.g. Krabbe disease, metochromatic leukodystrophy

Dopa responsive dystonia

Vitamin B12 deficiency / subacute combined degeneration of the cord

Abetalipoproteinaemia

Spinocereballar ataxias

HTLV-1 infection, tropical spastic paraparesis

Neurosyphilis

Neurolathyrism

Vitamin E deficiency

Arginase deficiency

MRI in hereditary spastic paraplegia may show a degree of spinal cord atrophy, but no other structural abnormalities of the central nervous system are usually apparent. Mild to moderate atrophy of the corpus callosum has been reported and several of these families have shown linkage to the SPG11 locus (Casali et al. 2004). There are occasional reports of atrophy of other intracranial structures and of cerebral hemisphere white matter lesions. Nerve conduction studies and electromyography are normal in the majority of cases of pure hereditary spastic paraplegia. Central motor conduction times have been reported to show either unrecordable, or delayed responses to the lower limbs, and usually normal values for the upper limbs (Pelosi et al. 1991; Schady et al. 1991). Somatosensory evoked potentials from the lower limbs have been reported to be small or absent (Pelosi et al. 1991; Aalfs et al. 1993).

The most useful molecular genetic test is screening for mutations in the spastin, SGP4 gene, which will be detected in approximately 40 per cent of patients with autosomal dominant hereditary spastic paraplegia. Such mutation screening is, however, costly and time consuming as mutations have been identified scattered throughout the spastin gene and therefore all 17 exons need to be analysed, at least in the index cases (Lindsey et al. 2000; McDermott et al. 2006). Mutations in atlastin, SPG3A, will be detected in another 10–15 per cent of families with autosomal dominant hereditary spastic paraplegia (Zhao et al. 2001) and the diagnostic yield is likely to be highest when targeted to patients with an age of onset of symptoms within the first decade of life.

To date 33 genetic loci associated with subtypes of hereditary spastic paraplegia have been identified and the 14 causative genes have been identified (Table 23.7). These causative genetic mutations may converge to cause disruption in several different cellular processes which may underlie hereditary spastic paraplegia and related disorders.

Autosomal dominant hereditary spastic paraplegia. To date 12 loci for autosomal dominant forms have been identified and the causative gene determined at 9 of these loci. The genes at SPG3A, SPG4, SPG6, SPG8, SPG10, SPG13, SPG17, SPG 31, and SPG 33 have been identified as atlastin, spastin, NIPA1, KIAA0196 or strumpellin, the kinesin heavy chain gene KIF5A, heat shock protein 60 (hsp60), seipin, REEP1, and ZFYVE27 respectively (Hazan et al. 1999; Zhao et al. 2001; Hansen et al. 2002; Reid et al. 2002; Rainier et al. 2003;Windpassinger et al. 2004; Mannan et al. 2006; Zuchner et al. 2006; Valdmanis et al. 2007;). The genes at the remaining loci SPG8, SPG9, SPG12, and SPG19 are as yet unknown.

Atlastin related hereditary spastic paraplegia; SPG3A. Hereditary spastic paraplegia linked to SPG3A on chromsome14q11.2 accounts for approximately 10 per cent of autosomal dominant cases (Zhao et al. 2001). Affected patients tend to have a pure phenotype, a young age of onset, and the presence of distal amyotrophy in the upper and lower limbs. Chronic neurogenic change on electromyography is another possible distinctive feature. Affected patients are sometimes misdiagnosed as having cerebral palsy. The SPG3A gene consists of 14 exons and encodes a protein of 588 amino acids which has conserved motifs for GTPase binding and hydrolysis and is structurally homologous to guanylate binding protein 1, a member of the dynamin family of large GTPases (Zhao et al. 2001). Its predicted structure indicates that it is likely to be an integral membrane protein, with two transmembrane domains (Zhu et al. 2003). Dynamins are a group of proteins known to be involved in vesicle trafficking, including the formation of clathrin coated vesicles from the plasma membrane and receptor mediated endocytosis; Golgi membrane dynamics; the maintenance and distribution of mitochondria and associate with cytoskeletal components including actin and the microtubule network. These functions are clearly important for neurotransmission, the action of neurotrophic factors, and axonal transport. The atlastin protein is localized most abundantly in the central nervous system, where it is enriched in pyramidal neurons, particularly in layer V of the cortex, including the motor cortex, and in the hippocampus (Zhu et al. 2003). At the subcellular level, it has recently been shown to co-localize with the Golgi apparatus (Zhu et al. 2003).

Most of the mutations in atlastin described to date are missense mutations which have been located in exons 4, 7, and 8. Of these only one, 650G>A, disrupts the GTPase motif directly (Muglia et al. 2002). It has been speculated that the other missense mutations may exert their pathogenic effect by introducing an altered secondary protein structure which disrupts GTPase activity by disturbing multimerization or protein–protein interactions. It will be of interest in the future to explore the effects of atlastin mutations on the structure and function of the Golgi apparatus, axonal transport and growth, as well as parameters such as endocyosis and vesicle trafficking, as perturbations of these functions could underlie the development of the distal axonopathy of hereditary spastic paraplegia.

Spastin related hereditary spastic paraplegia; SPG4. Spastin is made up of 17 exons mapping to chromosome 2p21-p22, and encodes a ubiquitously expressed 616 amino acid protein. SPG4 is the most frequent form of autosomal dominant hereditary spastic paraplegia, accounting for approximately 40 per cent of cases. Spastin related hereditary spastic paraplegia does not appear to have a distinctive clinical phenotype compared to other pure autosomal dominant types. In addition, there is no clear correlation between the type of spastin mutation identified and observed phenotype.

The age at onset for spastin related hereditary spastic paraplegia is highly variable as is the severity of the phenotype. Most affected individuals remain ambulant, but the clinical extremes of the phenotype range from asymptomatic patients with detectable lower limb pyramidal signs found only when clinically examined, about a quarter, to a minority of severely affected patients who become chairbound or bedridden. There is some evidence that disease progression and severity is worse in those with late onset disease and that the frequency of features such as paresis, amyotrophy, dorsal column involvement, and urinary disturbance increase with disease duration (Fonknechten et al. 2000). The majority of hereditary spastic paraplegia associated with spastin mutation is of a pure phenotype, but descriptions of more complicated phenotypes have emerged recently. Dementia or less severe disturbances of cognition complicating SPG4 has been described in several families and active progression of cognitive deterioration over a 3-year period in older patients with spastin related hereditary spastic paraplegia has been demonstrated (McGonagle et al. 2004). Epilepsy associated with spastin related hereditary spastic paraplegia has also been reported in several families (Mead et al. 2001) There are isolated reports of a variety of additional features complicating SPG4 including restless legs, myoclonus, atypical seizures, dysarthria, erectile dysfunction, severe constipation, ileus, and faecal incontinence.

Spastin shares homology with a large family of proteins known as the ATPases Associated with diverse cellular Activities, or AAA, which are involved in a range of cellular processes including cell cycle regulation, gene expression, organelle biogenesis, vesicle mediated protein transport, and as molecular chaperones co-operating in the assembly, function, and disassembly of protein complexes (Patel and Latterich 1998). All these AAA proteins share a common functional domain known as the AAA cassette which contains highly conserved motifs including Walker A, Walker B, and the AAA minimal consensus domain. Spastin has been shown to possess a microtubule interacting and trafficking domain and also has nuclear localization sequences (Ciccarelli et al. 2003). Spastin has particular homology with two other proteins, katanin and SKD1. Katanin is a microtubule severing protein and is involved in the dynamic regulation of the microtubule cytoskeleton throughout the cell cycle and SKD1 is an endosomal morphology and trafficking protein. There is evidence from cellular models generated in several laboratories that spastin may also be involved in regulating the microtubule cytoskeleton (Errico et al. 2002; McDermott et al. 2003a). Microtubules are dynamic polymers of alpha and beta tubulin. They perform essential roles, forming the mitotic spindle in dividing cells, acting as tracks for transport of various cellular cargoes including membranous organelles such as mitochondria and provide essential cytoskeletal support in all cells. Evidence to date suggests that spastin, like katanin, acts as a microtubule severing protein. It seems that missense mutations in spastin may lead to entrapment of the protein in a microtubule bound state. Thus, current knowledge indicates that spastin is involved in regulating microtubule dynamics and rearrangement both in proliferating and in post-mitotic cells. It is thought likely that spastin influences microtubule dynamics in growth cones, thus regulating the stability of axons and axonal transport. Long axons such as those of the corticospinal tracts are likely to be most dependent on fine regulation of the microtubule transport system by the action of spastin.

Of the mutations in the spastin gene published to date, approximately 11 per cent are nonsense mutations, 26 per cent frameshift with consequent premature termination codon, 28 per cent missense, and 35 per cent are splice site mutations. Recently, it has been reported that partial spastin deletions are relatively common and may account for approximately 18 per cent of cases of autosomal dominant hereditary spastic paraplegia (Beetz et al. 2006; Beetz et al. 2007). There is no particular ‘hot spot’ for mutation within the gene, with mutations throughout the length of the gene, making mutation screening a lengthy process. Most of the mutations are predicted to have a detrimental effect on the conserved AAA cassette. The broad mutational spectrum observed in spastin related hereditary spastic paraplegia initially suggested that axonal injury might be caused by a loss of function/haploinsufficiency effect. However, the demonstration of altered microtubule regulation in cells overexpressing missense spastin mutations suggests the possibility that a dominant negative pathogenic mechanism may be involved, at least in individuals with missense mutations. Spastin mutants with a missense mutation in the AAA cassette appear to bind constitutively to microtubules through the intact N terminal microtubule binding domain and could easily prevent the normal action of wild type spastin or otherwise block transport along the microtubule system. It is possible that more than one pathogenic mechanism may be involved, depending on the type of mutation. However, it seems likely that the pathogenesis of spastin related hereditary spastic paraplegia may be due to impairment of the fine regulation of the microtubule cytoskeleton in long axons.

Non-imprinted in Prader–Willi/ Angelman loci 1, or NIPA1 related hereditary spastic paraplegia; SPG6 is located within the 15q11 chromosomal region which is deleted in Prader-Willi and Angelman syndromes. Two families with autosomal dominant hereditary spastic paraplegia were initially described with the same T45R mutation in the NIPA1 gene (Rainier et al. 2003). This mutation disrupts an interspecies conserved amino acid. The function of NIPA1 was previously unknown, but it is strongly expressed in neuronal tissues and structural analysis of the encoded protein predicts that it possesses 9 transmembrane domains, suggesting that it may function as a membrane transporter or receptor (Rainier et al. 2003). It is possible therefore that the disease mechanism may involve altered signal transduction or small molecule transport. Recent evidence indicates that the NIPA1 protein functions as a magnesium transporter (Goytain et al. 2006). Individuals with Prader–Willi or Angelman syndromes with deletions including NIPA1 do not develop features of hereditary spastic paraplegia which suggests that the disease mechanism likely involves a dominant negative or gain of function rather than haploinsufficiency.

Strumpellin or KIAA0196 related hereditary spastic paraplegia; SPG8. Three mutations have been identified in families linked to the SPG8 locus (Valdmanis et al. 2007). The function of the encoded 1159 amino acid protein strumpellin is currently relatively unknown.

KIF5A related hereditary spastic paraplegia; SPG10. The kinesin heavy chain proteins KIF5A, KIF5B, and KIF5C are part of a multisubunit complex, kinesin-1, that acts as a microtubule motor involved in anterograde fast axonal transport. KIF5A has an exclusively neuronal expression, enriched in motor neurones (Hirokawa 1998). The heavy chain proteins represent the force producing subunit of kinesin, possessing a motor domain that interacts with the microtubule track and hydrolyses ATP. A single family has been described with pure, autosomal dominant hereditary spastic paraplegia associated with a missense mutation at an invariate asparagine residue, N256S, in the motor domain of the neuronal kinesin heavy chain protein KIF5A (Reid et al. 2002). The mutation is predicted to cause loss of function or a dominant negative effect on the neuronal kinesin I motor and is predicted to cause abnormal fast axonal transport of cargoes vital for the distal axon. A homologous mutation in Drosophila larvae results in ‘organelle jams’ within axons of motor neurons (Hurd et al. 1996). Recently a second autosomal dominant hereditary spastic paraplegia pedigree was described with a missense mutation, R280C, at an invariant arginine residue in exon 10, located in a region of the protein involved in microtubule binding activity (Fichera et al. 2004).

Heat shock protein 60 related hereditary spastic paraplegia; SPG13. The gene at the SPG13 locus has been identified as the mitochondrial chaperonin, heat shock protein 60. One family with a V72I substitution and autosomal dominant pure hereditary spastic paraplegia was described in 2002 (Hansen et al. 2002). The use of a complementation assay showed that wild type heat shock protein 60 and its co-chaperone heat shock protein 10, but not V72I heat shock protein 60 could support the growth of E. coli in which the homologous bacterial genes had been deleted (Hansen et al. 2002). Heat shock protein 60 is a mitochondrial protein which is upregulated for example in conditions of cellular stress, for example in the presence of an accumulation of unfolded proteins within the mitochondrial matrix (Zhao et al. 2002).

Seipin related hereditary spastic parapalegia or Silver syndrome, SPG17. Silver syndrome is a form of autosomal dominant hereditary spastic paraplegia in which spastic paraparesis is accompanied by atrophy of the hand muscles, especially the thenar muscles and sometimes the distal lower limb muscles (Silver 1966). Neurophysiological studies indicate lower motor neurone or motor root involvement in addition to the corticospinal tract dysfunction (Warner et al. 2004). Following evidence of linkage of some families to chromosome 11q12-q14, heterozygous missense mutations in the Berardinelli–Seip congenital lipodystrophy type 2 gene, BSCL2, were found in two families, one with Silver syndrome and one with hereditary distal motor neuropathy (Windpassinger et al. 2004). The encoded protein has been termed seipin and the two mutations result in amino acid substitutions, N88S and S90L, in an N-glycosylation motif. Seipin is an integral membrane protein of the endoplasmic reticulum and the disruption of glycosylation caused by the missense mutations appears to result in protein aggregation. The clinical phenotype of this genetic disorder has recently been expanded to include patients with predominantly lower limb amyotrophy and also to some patients without signs of corticospinal tract pathology (Irobi et al. 2004b). It had previously been demonstrated that null mutations affecting seipin caused autosomal recessive Berardinelli–Seip congenital lipodystrophy type 2, with the phenotype of near absence of adipose tissue from early childhood and severe insulin resistance. Currently it is not understood how mutant forms of seipin cause two such distinct disease states (Agarwal and Garg 2004).

Receptor expression-enhancing protein 1 related hereditary spastic paraplegia; SPG31. Recently mutations were identified in six autosomal dominant hereditary spastic paraplegia families linked to the SPG 31 locus on chromosome 2p12 (Zuchner et al. 2006). This represented 6.5 per cent of index cases with hereditary spastic paraplegia. Receptor expression-enhancing protein 1 is a widely expressed protein which localizes to mitochondria, although its precise function has not yet been elucidated.

ZFYVE27 related hereditary spastic paraplegia; SPG33. A recent report described a single German family with a mutation in ZFYVE27 (Mannan et al. 2006). The encoded protein is a novel member of the FYVE-finger family of proteins and is a specific spastin-binding protein.

Other autosomal dominant hereditary spastic paraplegia loci. The genes associated with SPG9, SGG12, and SPG19 have not yet been identified. Spastic paraparesis at the SPG9 has been described in one family in which the phenotype is complicated by bilateral cataracts, gastro-oesophageal reflux with vomiting, and amyotrophy (Seri et al. 1999). A further family has been described with cataracts, learning difficulties, and skeletal abnormalities (Slavotinek et al. 1996).

Autosomal recessive hereditary spastic paraplegia. Autosomal recessive hereditary spastic paraplegia is also clinically and genetically heterogeneous and is uncommon compared to the dominantly inherited forms. Autosomal recessive pedigrees have been linked to 15 genetic loci and include both pure and complicated phenotypes. The genes underlying three subtypes have been identified to date: SPG7, paraplegin (Casari et al. 1998); SPG20/Troyer syndrome, spartin (Patel et al. 2002); and SPG21, maspardin (Simpson et al. 2003). The genes associated with the other 12 autosomal recessive loci have not yet been identified.

Paraplegin related hereditary spastic paraplegia; SPG7. Patients with paraplegin mutations may have a pure or a complicated phenotype. In complicated cases the spastic paraparesis may be accompanied by a variety of other features including optic atrophy, cortical and cerebellar atrophy, dysphagia, dysarthria, distal amyotrophy, or sensorimotor neuropathy (Casari et al. 1998; McDermott et al. 2001). The SPG7 gene comprises 17 exons and the encoded protein, paraplegin, is a ubiquitously expressed nuclear encoded mitochondrial metalloprotease which is a member of the ATPase associated with diverse cellular activities or AAA, protein family. In yeast, Afg3p and Rca1p form a high molecular weight hetero-oligomeric complex in the inner mitochondrial membrane. From studies of homologous proteins in yeast, it was predicted that paraplegin would function by forming multimeric complexes which have proteolytic and chaperone-like functions in the mitochondria, essential for the normal assembly and turnover of respiratory chain complexes. Recent studies employing a paraplegin knockout mouse have shown that the first detectable abnormality is the appearance at 4 months of age of abnormal enlarged mitochondria in synaptic terminals in the grey matter of the lumbar spinal cord (Ferreirinha et al. 2004). These mitochondrial abnormalities correlate in time with the onset of a significant functional motor deficit and precede signs of axonal degeneration by several months. Clearly, mitochondrial dysfunction within axonal terminals could lead to problems with ATP generation, regulation of calcium homeostasis, and in free radical metabolism. The paraplegin knockout mice also develop accumulations of neurofilaments and organelles in swollen axons indicating abnormalities in anterograde transport. Retrograde transport is delayed in symptomatic mice and this could clearly contribute to the axonal degeneration by affecting the transport of mitochondria, the trafficking of endosomes, and the internalization of neurotrophic factors. Work with cellular models has demonstrated that paraplegin co-assembles with a homologous protein AFG3L2 into a high molecular weight complex within the inner mitochondrial membrane (Atorino et al. 2003). Using fibroblasts from SPG7 patients it was demonstrated that this complex is defective in the presence of paraplegin mutations, with a resultant decrease in complex 1 activity of the mitochondrial respiratory chain and increased sensitivity of the cells to oxidative stress. Several families have so far been described with paraplegin mutations. All the mutations described in some way affect the conserved AAA domain, either truncating the protein or by causing an in-frame deletion. Patients may show evidence of mitochondrial dysfunction on histochemical analysis of muscle (Casari et al. 1998; McDermott et al. 2001).

Spartin-related hereditary spastic paraplegia-Troyer syndrome; SPG 20. Troyer syndrome, named after the family in which it was first identified, was originally described in 1967 in an Old Order Amish community (Cross and McKusick 1967b), a genetically isolated population with high prevalence of autosomal recessive disorders. The cardinal clinical features include spastic paraparesis, pseudobulbar palsy, distal amyotrophy, mild developmental delay, and subtle skeletal abnormalities including short stature. Imaging studies may show cerebral white matter abnormalities and thinning of the corpus callosum (Proukakis et al. 2004). In 2002, a frameshift mutation, 1110delA, in SPG20 encoding spartin was found to be the genetic cause of this disorder (Patel et al. 2002). The SPG20 gene on chromosome 13q12.3 comprises nine exons and encodes a 72.7 kDa protein of 666 amino acids named spartin, which is ubiquitously expressed in adult tissues. Spartin shares homology with the other proteins, SNX15, VPS4, and Skd1, known to be involved in endosomal morphology and dynamics and protein trafficking, suggesting that spastin may serve a similar role. Spartin also shares homology with the N terminal region of spastin which forms the microtubule interacting and trafficking domain thought to be responsible for binding to microtubules. Thus, evidence to date indicates that spartin may function in the control of endosomal trafficking and/or microtubule dynamics (Crosby and Proukakis 2002). A very recent report described that spartin localizes to mitochondria (Lu et al. 2006).

Maspardin related hereditary spastic paraplegia—Mast syndrome; SPG 21. Mast syndrome is an autosomal recessive, complicated form of HSP accompanied by dementia, which is also found at relatively high frequency among the Old Order Amish community (Cross and McKusick 1967a). Clinical features may include developmental delay, pseudobulbar palsy, cerebellar and extrapyramidal dysfunction, as well as thinning of the corpus callosum, cerebral atrophy, and white matter demyelination on MR imaging of the brain. 14 affected patients were found to be homozygous for a single base pair, 601insA, insertion in the gene encoding the acid-cluster protein of 33 kDa, ACP33, which has been renamed maspardin (Simpson et al. 2003). This is a frameshift mutation which causes a truncated protein product which is likely to cause a loss of function effect. One study had reported the subcellular distribution of maspardin in a T cell-line (Zeitlman et al. 2001) where it localized to vesicles involved in the early endosome recycling pathway and to acidic organelles. Further work is required to define the localization and function of maspardin within neurons but current evidence indicates that maspardin may be involved in protein sorting and trafficking and that defective trafficking might be important in the pathogenesis of Mast syndrome.

X-linked forms of hereditary spastic paraplegia. These forms are relatively rare and are largely seen in paediatric practice. There are three X-linked loci: SPG1, SPG2, and SPG16. The genes involved at the SPG1 and two loci have been known for some time and the molecular mechanisms of disease are relatively well understood. The gene at SPG16 is not yet known.

L1 cell adhesion molecule related hereditary spastic paraplegia; SPG1. SPG1 is a rare developmental disorder of varying severity and patients usually have a complex phenotype with mental retardation and congenital musculoskeletal abnormalities, such as the absence of extensor hallucis longus, in addition to spastic paraparesis. Mutations in the L1 cell adhesion molecule gene, L1CAM, are also found in X-linked hydrocephalus, X-linked agenesis of the corpus callosum and the MASA syndrome of mental retardation, aphasia, shuffling gait and adducted thumbs. The diseases are now considered to be part of a clinical syndrome with the acronym CRASH: corpus callosum hypoplasia, retardation, adducted thumbs, spastic paraplegia, and hydrocephalus (Fransen et al. 1995). L1CAM is a cell surface, membrane associated glycoprotein, and a member of the immunoglobulin superfamily of proteins, which is expressed predominantly within neurons and Schwann cells. It is a protein with complex extracellular and intracellular interactions and has important functions in neuronal adhesion, axonal outgrowth, and pathfinding during central nervous system development. L1CAM plays an essential role in the correct formation of the corticospinal tract (Dahme et al. 1997; Demyanenko et al. 1999). In knockout mice the normal anatomy of the corticospinal tracts is disrupted with failure of decussation of corticospinal axons across the midline at the level of the pyramids (Castellani et al. 2000). L1CAM is a component of the Sema3A receptor complex and mice deficient in L1CAM fail to respond to directive signalling from Sema3A.

Proteolipid protein related hereditary spastic paraplegia; SPG2. The proteolipid protein 1 gene, PLP, encodes proteoipid protein 1 and its minor DM20 isoform. Both pure and complicated hereditary spastic paraplegia phenotypes may be observed in patients with mutations in proteolipid protein (Bonneau et al. 1993). Patients with a complicated phenotype may have a cerebellar syndrome, mental retardation, and optic atrophy in addition to spastic paraparesis. Mutations in the proteolipid protein gene also cause Pelizaeus–Merzbacher disease, a severe dysmyelinating syndrome characterized by nystagmus, ataxia, spasticity, abnormal movements, optic atrophy, and microcephaly, usually resulting in death during adolescence (Section 10.2.10). The phenotype of disease caused by mutations in the proteolipid protein gene can be considered as a continuous spectrum with milder SPG2 at one end and the more severe Pelizaeus–Merzbacher disease at the other. Proteolipid protein is the major myelin protein of the central nervous system, accounting for approximately 50 per cent of total myelin protein in the adult brain. DM20 is an alternatively spliced form, which lacks 35 amino acid residues. The function of the proteolipid protein and DM20 proteins has not been established with certainty, but they are considered likely to have an important function in stabilizing the structure of central nervous system myelin by forming the intraperiod line. It is possible that the two protein isoforms have different functions. The DM20 isoform appears earlier during development and is thought to play a role in glial cell development, while the proteolipid protein isoform is expressed later and may play a role in myelin assembly and maintenance. Mutations in the PLP gene that do not affect the the DM20 isoform are associated with the milder Pelizaeus–Merzbacher disease phenotype or SPG2. Conversely mutations which reduce the level of the DM20 isoform are associated with the more severe Pelizaeus–Merzbacher disease phenotypes (Griffiths et al. 1998a). Plp knockout mice develop normal myelin sheaths despite lack of PLP/DM20, but subsequently develop a severe axonopathy (Griffiths et al. 1998b). It was recently demonstrated that absence of PLP/DM20 in oligodendrocytes results in early impairment of fast axonal transport and multifocal accumulations of membranous organelles, in a mouse model with a null mutation in PLP (Edgar et al. 2004). Axonal degenerative changes were found to be concentrated at the distal regions of long axons.

So far, increased knowledge of the molecular basis of hereditary spastic paraplegia has had little impact on clinical practice. At present symptomatic treatments can be used to alleviate spasticity, bladder dysfunction, and the lower limb discomfort which can be troublesome features of hereditary spastic paraplegia. Neuroprotective therapies are not yet available to ameliorate the distal axonopathy.

Kennedy’s disease, or spinobulbar muscular atrophy is an X-linked degenerative disorder of lower motor neurones (Kennedy et al. 1968). Initial symptoms consist of hand tremor, fasciculations, and muscle cramps, followed by progressive weakness and atrophy of limb and bulbar musculature (Sinnreich and Klein 2002; Lee et al. 2005). Limb muscle weakness is predominantly proximal in distribution and mainly affects the lower limbs. There are no clinical signs of upper motor neurone dysfunction and tendon reflexes are reduced or absent. Weakness of the lower facial and tongue muscles causes dysarthria and jaw weakness may cause the mouth to hang open. Pharyngeal involvement causing dysphagia, and respiratory muscle weakness causing breathlessness, are less common. Peri-oral fasciculations causing ‘quivering’ of the chin is a characteristic feature. Patients frequently have evidence of mild androgen insensitivity with gynaecomastia, testicular atrophy, oligospermia, and erectile dysfunction. The disease course is much more slowly progressive compared to amyotrophic lateral sclerosis and typically life expectancy is not reduced, though a proportion of patients may die from the effects of respiratory muscle weakness (Atsuta et al. 2006). Patients may become wheelchair dependent over 2–3 decades, although some remain ambulatory until late in life. Sensory symptoms are uncommon and mild. However there is evidence that neurodegeneration is not exclusive to motor neurones. Mild distal sensory loss is frequently present in the lower limbs, and sensory nerve conduction studies show decreased or absent sensory nerve potentials in keeping with depletion of dorsal root ganglion cells demonstrated pathologically (Ferrante and Wilbourn 1997). Heterozygous female carriers of spinobulbar musclar atrophy may show mild clinical manifestations of the disease such as muscle cramps, mild facial weakness, and fasciculations, and neurophysiological studies showing mild changes of chronic denervation (Sobue et al. 1993).

Kennedy’s disease is often misdiagnosed initially as amyotrophic lateral sclerosis. Though rare, it is important not to miss the diagnosis because of the genetic implications for the family and the comparatively more benign disease course. The diagnosis should be considered in any male patient with a phenotype of a pure lower motor neurone disorder, particularly when the disease course is relatively indolent, gynaecomastia is present, or when there is evidence of a mild concomitant sensory neuropathy. DNA examination for the CAG repeat expansion in exon 1 of the androgen receptor gene is diagnostic.

Spinobulbar muscular atrophy was linked to chromosome Xq12-21 in 1986 (Fischbeck et al. 1986). The disease-causing mutation was reported in 1991 identifying expansion of a trinucleotide CAG repeat in the androgen receptor gene from a normal length of 17–26 repeats, to a disease associated length of 40–52 repeats (La Spada et al. 1991). This CAG repeat region encodes a long tract of glutamine residues, beginning at amino acid 58. Disease severity and age of onset show some correlation with the size of the repeat expansion (Doyu et al. 1992). Patients with minimally expanded polyglutamine tracts in the androgen receptor gene have been reported to have unusual clinical manifestations, including essential tremor (Kaneko et al. 1993a), hypertrophic cardiomyopathy (Kaneko et al. 1993b), and very late onset of muscular weakness (Doyu et al. 1993). Spinobulbar muscular atrophy is one of the triplet repeat expansion disorders. In all these disorders, neurodegeneration occurs when the poly(Q) tract reaches a critical length of about 40 repeats and there tends to be a similar age of onset and rate of progression of disease, which correlate with the length of the poly(Q) tract (Zoghbi and Orr 2000). All are progressive neurological disorders that affect only a subset of neurones, despite ubiquitous expression of the expanded protein.

The androgen receptor is a nuclear receptor in the steroid receptor superfamily. The protein, encoded by eight exons, contains three functional domains: a carboxy-terminal hormone-binding domain, a DNA-binding domain, and an amino-terminal transactivation domain, which contains the polyglutamine tract. The androgen receptor is phosphorylated and bound to heat shock proteins in the cytoplasm. On ligand binding, it is transported to the nucleus, where it binds to DNA, and acts as a transcription factor. Expansion of the polyglutamine tract results in reduced target gene transactivation which may account for the features of decreased androgen sensitivity (Lieberman et al. 2002). However, complete loss of androgen receptor function, in individuals with testicular feminization syndrome, does not lead to motor neurone degeneration. Reduction in androgen receptor function does not therefore appear to be the major mechanism leading to motor neurone degeneration in spinobulbar muscular atrophy, and it is thought that neurodegeneration occurs through a toxic gain of function of the altered protein. There is evidence that polyglutamine repeats themselves are neurotoxic whilst the protein in which it is expressed is likely to modify the specific pattern of neurodegeneration seen in each trinucleotide repeat disorder (Ordway et al. 1997).

A transgenic mouse model of Kennedy’s disease has been developed with a full-length androgen receptor and very long repeat expansions, which phenotypically reproduces spinobulbar muscular atrophy (Katsuno et al. 2002). There is evidence that toxicity of the mutant androgen receptor is ligand dependent, as it only occurs with the higher androgen levels present in males. Castration of the transgenic male mice reduces symptom severity, whilst treatment of transgenic females with exogenous testosterone worsens their phenotype (Katsuno et al. 2002). Leuprorelin, an LHRH agonist that reduces testosterone release from the testis, inhibits nuclear accumulation of mutant androgen receptor protein, resulting in rescue of the motor dysfunction in male transgenic mice (Katsuno et al. 2004). Human clinical trials of leuprorelin in spinobulbar muscular atrophy patients have commenced.

Several potential mechanisms for the toxic gain of function of the expanded androgen receptor protein have been proposed. Misfolding of the polyQ expanded proteins in spinobulbar muscular atrophy and other polyglutamine diseases is thought to lead to formation of aggregates, which are seen as nuclear inclusions. Nuclear inclusions containing the amino-terminal epitopes of the mutant androgen receptor are found within motor neurones and certain non-neuronal tissues in patients with spinobulbar muscular atrophy (Li et al. 1998) and in cell culture and animal models of the disease. The role of protein aggregation in the pathogenesis of neurodegeneration is, however, uncertain. The presence of ubiquitin, proteasomes, and molecular chaperones within nuclear inclusions in spinobulbar muscular atrophy implicates protein misfolding in disease pathogenesis. There is evidence that polyglutamine proteins may directly impair the ubiquitin–proteasome system, possibly by exceeding the capacity of proteasomes or sequestering molecular chaperones, which would then be unavailable to degrade other substrates (Bence et al. 2001). Ubiquitin-dependent proteolysis has a central role in regulating fundamental cellular events, such as cell division and apoptosis, and represents a potential mechanism linking protein aggregation to cellular dysregulation and cell death.

Nuclear localization of mutant androgen receptor appears to be a requirement for neurotoxicity (Katsuno et al. 2002). Nuclear accumulation of mutant polyglutamines may cause toxicity through interaction with transcription factors, with resultant disruption of gene transcription. Down-regulation of gene expression seems to be an early event in polyglutamine disease pathogenesis. In the case of spinobulbar muscular atrophy, interest has focussed on the CREB binding protein, CBP, which is sequestered by the mutant polyglutamine (McCampbell et al. 2000). CBP is a transcriptional coactivator that possesses histone acetyltransferase activity. Hyperacetylation of histones marks transcriptionally active regions of chromatin. As CBP is present in a functionally limiting level in cells, its sequestration by mutant polyglutamine could have major effects on gene expression. In neurons, CBP is a key component of neurotrophic factor signalling pathways, including that of vascular endothelial growth factor, VEGF. Transgenic mice that overexpress mutant human androgen receptor have reduced spinal cord VEGF164 protein levels, even in presymptomatic stages, and polyglutamine-expanded androgen receptor has been shown to interfere with CBP-mediated transcription of VEGF (Sopher et al. 2004). Reversal of the histone acetylation defect caused by CBP sequestration, using histone deacetylase inhibitor drugs has been shown to reduce polyglutamine-associated cell death in both Drosophila and mouse models of Huntington’s disease (Steffan et al. 2001; McCampbell et al. 2001).

The mutant androgen receptor protein may also adversely affect axonal transport. A motor neurone cell line transfected with mutant androgen receptor, develops protein aggregates which affect the distribution of the motor protein kinesin, and consequently the distribution of mitochondria. This suggests that protein aggregation within motor neurones in Kennedy’s disease disrupts kinesin-mediated fast axonal transport (Piccioni et al. 2002).

The Brown–Vialetto–van Laere syndrome is a rare disorder of unknown aetiology characterized by progressive weakness of bulbar muscle groups associated with sensorineural deafness (Brucher et al. 1981; Hawkins et al. 1990; Sathasivam et al. 2000). In most cases progressive deafness is the initial symptom followed by involvement of the lower motor cranial nerves (Fig. 23.7). Lower motor neurone and less commonly upper motor neurone signs in the limbs may develop. The disorder may occur sporadically, but in some cases is familial, usually with autosomal recessive inheritance. The age of onset of the first symptom varies from infancy to the third decade. A period of worsening may be associated with intercurrent events such as an infection.

 A patient with Brown–Vialetto–van Laere syndrome showing bilateral facial weakness (A) and wasting of the tongue (B).
Fig. 23.7

A patient with Brown–Vialetto–van Laere syndrome showing bilateral facial weakness (A) and wasting of the tongue (B).

Neurophysiological examination reveals changes of chronic or active denervation in limb and bulbar muscles with normal nerve conduction velocities. Detectable hyperintensity of brainstem nuclei on MRI has been recently described in some patients (Koul et al. 2006). The clinical course of the condition is variable; some patients deteriorate progressively from the outset, others have abrupt periods of deterioration interspersed with periods of stability. Approximately half of patients reported in the literature survive more than 10 years. Management of patients at the present time consists of symptomatic treatment and supportive care including gastrostomy feeding and respiratory support measures.

Fazio–Londe disease is a rare, and presumed to be inherited either autosomal recessively or dominantly, form of progressive motor neurone degeneration with onset in infancy (McShane et al. 1992). The disorder develops in children within the first 5 years of life. Stridor is a prominent early symptom, often leading to an initial diagnosis of croup. Progressive bulbar paralysis then develops, with dysarthria and dysphagia. Typically, over the next 2 years the limb muscles become involved and respiratory failure develops resulting in death. Occasional cases of later onset and longer survival have been described. Fazio–Londe syndrome and Brown–Vialetto–van Laere syndrome may overlap, as shown in one family in which four siblings born to consanguineous parents were variably affected by pontobulbar weakness, deafness, and limb weakness (Dipti et al. 2005). Two siblings with the phenotype of Fazio–Londe disease have been described who had a mitochondrial respiratory chain defect (Roeleveld-Versteegh et al. 2004).

The development of motor system disorders has been associated with dietary dependence on food plants with neurotoxic potential, notably the grass pea or chickling pea Lathyrus sativus and cassava Manihot esculenta Crantz, in specific geographical regions of the world. Dietary reliance on the grass pea or on insufficiently processed cassava has resulted in outbreaks of neurolathyrism, and konzo or neurocassivism, respectively. These are self-limiting neurodegenerative disorders confined to the upper motor neurone system. Investigation of epidemics of lathyrism and konzo has indicated that individual susceptibility to the toxic effects of these plants varies with gender, age, nutritional state, level of physical exercise, as well as the toxin content of the plants, methods of preparation of staple food, and duration of dietary exposure. Epidemics of both disorders tend to occur when adverse environmental conditions force heavy dietary reliance on the grass pea and cassava, which are plants relatively resistant to drought and pest attack. Despite the clinical similarities of the two neurotoxic disorders, outbreaks of these diseases have occurred in two distinct non-overlapping geographical areas of the world. Lathyrism outbreaks have occurred in the Indian subcontinent, China, and in Ethiopia and Eritrea on the Horn of Africa as well as parts of Europe. By contrast konzo has been reported mainly from sub-Saharan Africa (Rosling and Tylleskar 1995; Rosling and Tylleskar 2000).

The common clinical picture in both disorders is of a symmetrical spastic paraparesis. In the prodromal phase subjects affected by lathyrism or konzo complain of leg stiffness, weakness, and cramping. Tremor is common and acute reversible sensory symptoms are often reported. Bladder involvement is not common. In mild cases spasticity of the legs is only revealed when the subject is asked to run; in severe cases the individual may become bedridden. In time, severely affected patients may show disuse muscle atrophy. Sensory examination, mentation, cerebellar and cranial nerve function are usually normal. Konzo, the cassava related disorder, is potentially more serious and may cause a spastic tetraparesis as well as pseudobulbar signs (Tshala-Katumbay et al. 2001). Severe cases of konzo may also develop visual impairment and bilateral optic neuropathy (Mwanza et al. 2003). The term konzo means ‘tied legs’ in the Kiyaka language. There is evidence for another cassava associated neurological disorder which occurs in older subjects with a chronic high intake of incompletely detoxified cassava (Osuntokum 1981). This consists of a slowly evolving ataxic neuropathy with or without evident upper motor neurone signs, which may be accompanied by visual and sensorineural hearing deficits.

The two diseases show differential patterns of gender susceptibility. Lathyrism tends to affect young males more commonly and more severely than females (Spencer et al. 1984). Konzo appears to affect males and females similarly, while women of child bearing age are more susceptible than men of the same age (Tylleskar et al. 1993). Excessive physical activity is often reported at the outset of disease and may be a predisposing factor that stresses the motor system and promotes susceptibility of the cortical motor neurones.

Electrophysiological examination in both disorders indicates marked dysfunction of the pyramidal tracts with absent responses in the lower limbs following magnetic or transcranial magnetic stimulation of the motor cortex or prolonged central motor conduction times (Hugon et al. 1990, Tshala Katumbay et al. 2002). Most subjects have normal peripheral motor and sensory conduction velocities and electromyography. Somatosensory evoked response studies indicate that subclinical involvement of sensory pathways is relatively common. A study using MRI in two Tanzanian patients with konzo revealed no abnormalities (Tylleskar et al. 1993). Imaging studies are not available for patients with lathyrism. Pathological studies are sparse or lacking. A study of the brain in one individual who developed lathyrism 31 years prior to his death revealed loss and shrinkage of pyramidal neurons in the upper part of the precentral gyrus (Filiminoff 1926). A few studies of the spinal cord in cases of lathyrism have shown predominantly distal and symmetrical degeneration of lateral and ventral corticospinal tracts, sometimes accompanied by distal degeneration of the spinocerebellar and gracile tracts (Hirano et al. 1976; Striefler et al. 1977).

The molecular mechanisms underlying these two neurotoxic disorders have still to be elucidated. β-N-oxalylamino-L-alanine, or BOAA, appears to be the main neurotoxic compound in the grass pea and is considered the likely cause of human lathyrism. The mechanism of neurotoxicity appears to be excitotoxic, involving excessive glutamatergic neuronal stimulation. BOAA is a potent agonist at the AMPA subtype of glutamate receptor and an inhibitor of glutamate uptake (Spencer 1999) and recent evidence indicates that activation of group I metabotropic glutamate receptors may also play a part in the toxic effects (Kusama-Eguchi et al. 2004). Other studies have provided evidence that BOAA may disrupt mitochondrial function by triggering glutathione loss and protein thiol oxidation (Ravindranath 2002). At present, there is no robust animal model of lathyrism that develops features of cortical motor neurone degeneration.

In konzo the disease pathogenesis is not properly understood and again there is no animal model of the condition. Epidemiological studies have consistently shown a link with dietary intake of inadequately processed bitter cassava, combined with a low protein intake (Rosling and Tylleskar 2000). Bitter varieties of cassava contain large amounts of cyanogenic glucosides, linamarin, and lotaustralin, and the levels of these poisons depend on the prevalent environmental conditions. Weeks or months of dependency on incompletely detoxified cassava, combined with low intake of proteins which normally provide the source of sulphur amino acids to convert cyanide to thiocyanate, leads to outbreaks of konzo. The mechanism by which cyanogenic toxins damage the upper motor neurone system has not been established with certainty: hypotheses include mitochondrial inhibition; modulation of the glutamatergic neurotransmitter system (Spencer 1999); and carbamoylation of proteins in the nervous system (Mellado et al. 1982). The cyanide metabolite cyanate is considered the likely aetiological factor in the cassava induced ataxic (myelo)neuropathy (Kogure et al. 1975).

There is no effective treatment for these persistently disabling disorders. Once the acute or subacute onset of the disease has passed, the disability remains unchanged and irreversible. Little benefit has been reported from centrally acting spasmolytic agents. The role of therapies such as botulinum toxin injection and physiotherapy to reduce muscle spasm and prevent joint contractures has not been reported. However, both lathyrism and konzo can be prevented by education, modifying food preparation, or changing dietary practice, and international networks have been established to aim at preventing outbreaks of both lathyrism and konzo.

In the 1950s a high incidence geographical focus of amyotrophic lateral sclerosis was identified on the Western Pacific island of Guam, where the indigenous Chamorro people had a rate of the disease of approximately 50–100 times that of Western populations (Waring et al. 2004). Subsequently a unique neurodegenerative condition amongst the same population, which was given the term Parkinsonism–dementia complex was described (Hirano et al. 1961). Affected individuals developed severe dementia, and rigid akinetic Parkinsonism with marked postural deformities. Hyperreflexia and muscle atrophy affecting mainly the distal extremities were frequently detected. Familial clustering of both disorders was observed. The occurrence of Parkinsonism–dementia complex and amyotrophic lateral sclerosis in a geographically isolated population in similar age groups, and with overlap in symptomatology and familial aggregation led to the suspicion that the two diseases may have a common cause (Murakami 1999). Pathologically, Guamanian amyotrophic lateral sclerosis–Parkinsonism–dementia complex appears to be a tauopathy, characterized by the formation of neurofibrillary tangles. Variation in the anatomical distribution of pathology accounts for the varying clinical presentations, with tangles predominantly seen in the motor system in Guamanian amyotrophic lateral sclerosis (Ince and Codd 2005).

The cause of Parkinsonism–dementia complex and Guamanian amyotrophic lateral sclerosis is unknown. A genetic cause has not been found despite apparent familial clustering of the diseases. Several observations support an environmental aetiology, including the fact that Filipino immigrants are susceptible to the condition, and that there is a trend for increasing age of onset and decreasing disease incidence over time (Galasko et al. 2002). The leading hypothesis relates to ingestion of an environmental toxin present in the seed of the false sago palm, Cycas circinalis (Ince and Codd 2005). Beta-methylaminoalanine, BMAA, is one of several chemicals with neurotoxic properties present in the seeds of cycad plants which are used by the native Chamorros for food and medicine. BMAA can selectively injure motor neurones via AMPA/kainate receptor activation followed by increased intracellular calcium and free radical generation (Rao et al. 2006). BMAA toxicity seemed unlikely as a cause of Guamanian amyotrophic lateral sclerosis when studies showed that the Chamorro practice of flour preparation from cycad seeds largely removed the toxin and that neurotoxic effects in primates were only observed following massive exposure to BMAA. However, the cycad hypothesis has emerged recently based on a new understanding of Chamorro food practices, a cyanobacterial origin of BMAA in cycad tissue and a possible mechanism for biomagnification of this neurotoxin in Guam ecosystem (Ince and Codd 2005). Free living cyanobacteria produce high levels of BMAA in the root system of cycads and the toxin is then concentrated in the seed tissues. The traditional Chamorro diet includes the prized delicacy of the fruit bat, which feeds on cycad seed components and reportedly bioaccumulates BMAA (Cox et al. 2003). Plant and animal proteins provide a previously unrecognized reservoir for the slow release of the BMAA toxin. Patients with amyotrophic lateral sclerosis–Parkinsonism–dementia complex have been reported to show elevated levels of BMAA in post-mortem brain tissue (Cox et al. 2003). Further investigation of this aetiological hypothesis and indeed of the potential role of cyanobacterial toxicity in sporadic neurodegenerative diseases is ongoing.

Approximately 5 per cent of patients undergoing radiotherapy for breast cancer develop brachial plexopathy, but in addition to motor problems, sensory change is usually prominent (Olsen et al. 1993; Jaeckle 2004). Neurophysiological examination may reveal myokymia and motor nerve conduction block (Esteban and Traba 1993). Delayed progressive bulbar dysfunction with myokymia has also been described in some patients following radiotherapy (Glenn and Ross 2000). Post-irradiation myelopathy can produce a relatively pure lower motor neurone syndrome (Lamy et al. 1991).

A predominantly motor disorder can develop in the lower limbs following radiotherapy involving exposure of the lumbar spinal canal as part of therapy for testicular tumours, lymphoma, or other neoplasms (Section 5.9.5). The latency between radiotherapy and onset of symptoms varies between a few months and several decades. The lower limb weakness is often distal and asymmetrical and associated with loss of reflexes. Wasting may not be apparent even in weak muscles, indicating that conduction block may contribute to weakness, although this is difficult to demonstrate. In most patients the leg weakness gradually progresses and may eventually result in severe disability, though there may be periods of stabilization of symptoms. Patients may eventually develop sensory symptoms, but objective sensory signs are usually absent and sensory nerve action potentials are normal (Bowen et al. 1996). Sphincter and sexual function are usually relatively well preserved initially. Attempted therapeutic intervention with steroids or intravenous immunoglobulin has not shown any convincing benefit. MRI with gadolinium may show nodular enhancement of the conus and cauda equina (Bowen et al. 1996; Hsia et al. 2003). The CSF protein may be normal or elevated. Myokymia may be noted on electromyography. Microscopic examination has shown a vasculopathy of the proximal spinal nerve roots, with preservation of motor neurone cell bodies and spinal cord architecture (Bowen et al. 1996). With improved safety of radiotherapy regimes and the substitution of chemotherapy for radiotherapy in the treatment of testicular cancer, it is expected that the incidence of this clinical problem will decrease over time

Clinicians use several terms, including myokymia, neuromyotonia, Isaac’s syndrome, and cramp fasciculation syndrome to describe the motor manifestations of peripheral nerve hyperexcitability (Section 24.10.4). Neuromyotonia is a rare syndrome of spontaneous and continuous muscle fibre activity reflecting a peripheral nerve hyperexcitability disorder (Maddison 2006). This results in stiffness and cramping of muscles, often worse following exercise, though muscle power is frequently normal. Neuromyotonia can occur as a feature of a heterogeneous group of underlying disorders. The majority of cases are acquired and some of these are associated with an underlying peripheral neuropathy, myasthenia gravis, or the administration of penicillamine (Hart et al. 2002). Up to 20 per cent of cases may be associated with an underlying malignancy, usually thymoma or small cell lung cancer (Caress et al. 1997). Neuromyotonia can also occasionally be found in patients with hereditary neuropathies (Hahn et al. 1991). In acquired neuromyotonia, voltage-gated potassium channel, or VGKC, antibodies indicative of an auto-immune pathogenesis may be found in approximately 40 per cent of patients (Hart et al. 1997). There is evidence that these antibodies may reduce K+ currents by cross-linking of potassium channels (Tomimitsu et al. 2004).

Neurophysiological assessment allows neuromyotonia to be distinguished from true myotonia. Electromyography features include spontaneous, continuous, irregularly occurring doublet or multiplet single motor unit, or partial motor unit, high intraburst frequency of 30–300 Hz (Fig. 23.8). Patients do not have detectable central neurophysiological abnormalities and motor cortex excitability parameters, and central motor conduction times are normal (Maddison et al. 2006). The pathogenic abnormality is thought to lie within the terminal branches of the peripheral motor nerves. Symptoms of neuromyotonia may improve substantially with carbamazepine or phenytoin therapy and patients may respond to intravenous immunoglobulin therapy or plasma exchange.

 Neuromyotonia. The upper traces are of a 25-s continuous needle electromyography recording from medial gastrocnemius muscle in a patient with acquired neuromyotonia. Sections of this are shown at faster time–bases below. Motor units are seen to fire spontaneously and irregularly as doublets (B), triplets (A) and multiplets (C), with intraburst frequencies of up to 120 Hz. The prolonged discharge seen in the middle of the recording consists of rapidly firing triplets of more than one motor unit (D). (Courtesy of Dr Paul Maddison.)
Fig. 23.8

Neuromyotonia. The upper traces are of a 25-s continuous needle electromyography recording from medial gastrocnemius muscle in a patient with acquired neuromyotonia. Sections of this are shown at faster time–bases below. Motor units are seen to fire spontaneously and irregularly as doublets (B), triplets (A) and multiplets (C), with intraburst frequencies of up to 120 Hz. The prolonged discharge seen in the middle of the recording consists of rapidly firing triplets of more than one motor unit (D). (Courtesy of Dr Paul Maddison.)

The differential diagnosis of neuromyotonia includes true myotonic disorders, myokymia which is restricted to muscle innervated by injured motor axons, the stiff person syndrome (Section 23.7.2), and the benign cramp fasciculation syndrome.

Stiff person syndrome is a rare distressing neurological disorder first described in 1956 (Moersch and Woltman 1956) (Sections 38.3.4 and 40.10.3). The symptoms often begin with tightness or stiffness of the trunk muscles which will usually spread to involve the proximal muscles of the limbs. Persistent contraction of the axial muscles leads to truncal rigidity, a characteristic lumbar hyperlordosis, and restriction of movement of the hips and spine. Superimposed on this background rigidity, the patient will often be affected by painful muscle spasms which may be precipitated by sudden movement, noise, tactile stimulation, or emotional upset. Specific phobia is a frequent non-motor symptom of stiff person syndrome (Henningsen and Meinck 2003). Neurophysiological examination reveals motor unit firing at rest simultaneously from agonist and antagonsist muscles. The spontaneous muscle activity is of central rather than peripheral origin, related to release of polysynaptic spinal and brainstem reflexes (Thompson 2001). Up to 80 per cent of patients with classical stiff person syndrome have autotantibodies in the serum and CSF to glutamic acid decarboxylase, GAD, a rate limiting enzyme responsible for the synthesis of gamma-aminobutyric acid, GABA, and concentrated in GABAergic nerve terminals and in pancreatic β cells (Rakocevic et al. 2004). A high proportion of patients have coexisting diabetes mellitus or other autoimmune diseases. More recently antibodies to GABA A-receptor-associated-protein have been found in up to 70 per cent of patients with stiff person syndrome, with experimental evidence that this may inhibit the surface expression of the GABA A receptor and the normal functioning of GABAergic neurotransmission (Raju et al. 2006). In patients with classical stiff person syndrome there is a paucity of neuropathological abnormalities, suggesting that functional impairment of inhibitory neurotransmitter circuitry is sufficient to cause disease without overt structural damage. Imaging studies are usually normal in classical stiff person syndrome, but magnetic resonance spectroscopy has shown reduced levels of GABA in specific brain regions (Levy et al. 2005) and transcranial magnetic stimulation has revealed significantly enhanced motor cortex excitability (Koerner et al. 2004).

Several variants of the stiff person syndrome have been described (Shaw 1999). Progressive encephalomyelitis with rigidity is usually a paraneoplastic disorder which may present with similar clinical features. However, the clinical course is usually relentlessly progressive, often resulting in death within a few months. Histopathological examination reveals widespread perivascular lymphocytic cuffing and infiltration, with neuronal loss in the lower brainstem and spinal cord, especially in the spinal grey matter (Whitely et al. 1976). A second variant is the jerking stiff person syndrome, in which myoclonic jerking is a prominent additional feature (Leigh et al. 1980). A third variant, in which antibodies to the synaptic vesicle protein amphiphysin I are detected, has been described in association with breast cancer (Folli et al. 1993). Barker and colleagues drew attention to a fourth clinical sub-type, the stiff limb syndrome (Barker et al. 1998). Affected individuals present with rigidity, abnormal fixed posturing, and painful spasms usually of the distal lower limb and sparing the trunk. Brainstem abnormalities and sphincter dysfunction may develop later in the disease course. This focal variant is less likely to be associated with detectable autoantibodies, or to respond well to therapy and is more likely to lead to loss of ambulation. Recently a series of patients with stiff person syndrome and elevated anti-GAD antibodies in the CSF has been reported who also had cerebellar disease, gait ataxia, dysarthria, and oculomotor abnormalities (Rakocevic et al. 2006).

The rarity of stiff person syndrome limits the feasibility of controlled clinical trials, but the available evidence indicates that drugs which increase cortical and spinal inhibition or enhance GABA such as benzodiazepines, baclofen, valproate, gabapentin, tiagabine, and immunomodulatory therapies such as prednisolone, intravenous immunolglobulin, and plasmapheresis are effective therapies.

Aalfs
CM, Koelman JH, Posthumus Meyjes FE et al. (
1993
).
Posterior tibial and sural nerve somatosensory evoked potentials: a study in spastic paraparesis and spinal cord lesions.
 
Electroencephalogr Clin Neurophysiol
, 89, 437–41.

Abe
K, Fujimura H, Tatsumi, C et al. (
1995
).
Eyelid “apraxia” in patients with motor neuron disease.
 
J Neurol Neurosurg Psychiatry
, 59, 629–32.

Agarwal
AK, Garg A (
2004
).
Sepin: a mysterious protein.
 
Trends Mol Med
, 10, 440–4.

Aguirre
T, Van Den Bosch L, Goetschalckx K et al. (
1998
).
Increased sensitivity of fibroblasts from amyotrophic lateral sclerosis patients to oxidative stress.
 
Ann Neurol
, 43, 452–7.

Almer
G, Teismann P, Stevic Z et al. (
2002
).
Increased levels of the pro-inflammatory prostaglandin PGE2 in CSF from ALS patients.
 
Neurology
, 58, 1277–9.

Andersen
PM, Forsgren L, Binzer, M et al. (
1996
).
Autosomal recessive adult-onset amyotrophic lateral sclerosis associated with homozygosity for Asp90Ala CuZn-superoxide dismutase mutation. A clinical and genealogical study of 36 patients.
 
Brain
, 119, 1153–72.

Antonellis
A, Ellsworth RE, Sambuughin N et al. (
2003
).
Glycyl tRNA synthetase mutations in Charcot-Marie-Tooth disease type 2D and distal spinal muscular atrophy type V.
 
Am J Hum Genet
, 72, 1293–9.

Araki
K, Ueda Y, Michinaka C et al. (
1989
).
An autopsy case of juvenile muscular atrophy of unilateral upper extremity (Hirayama’s disease).
 
Nippon Naika Gakkai Zasshi
, 78, 674–5.

Armon
C (
2003
).
An evidence-based medicine approach to the evaluation of the role of exogenous risk factors in sporadic amyotrophic lateral sclerosis.
 
Neuroepidemiology
, 22, 217–28.

Atorino
L, Silvestri L, Koppen M et al. (
2003
).
Loss of m-AAA protease in mitochondria causes complex I deficiency and increased sensitivity to oxidative stress in hereditary spastic paraplegia.
 
J Cell Biol
, 163, 777–87.

Atsuta
N, Watanabe H, Ito M et al. (
2006
).
Natural history of spinal and bulbar muscular atrophy (SBMA): a study of 223 Japanese patients.
 
Brain
, 129, 1446–55.

Auer-Grumbach
M, Fazekas F, Radner H et al. (
1999
).
Troyer syndrome: a combination of central brain abnormality and motor neuron disease?
 
J Neurol
, 246, 556–61.

Azzouz
M, Ralph GS, Storkebaum E et al. (
2004
).
VEGF delivery with retrogradely transported lentivector prolongs survival in a mouse ALS model.
 
Nature
, 429, 413–7.

Bak
TH, O’Donovan DG, Xuereb JH et al. (
2001
).
Selective impairment of verb processing associated with pathological changes in Brodmann areas 44 and 45 in the motor neurone disease-dementia-aphasia syndrome.
 
Brain
, 124, 103–20.

Barker
RA, Revesz T, Thom M et al. (
1998
).
Review of 23 patients affected by the stiff man syndrome: clinical subdivision into stiff trunk (man) syndrome, stiff limb syndrome, and progressive encephalomyelitis with rigidity.
 
J Neurol Neurosurg Psychiatry
, 65, 633–40.

Beal
MF (
2000
).
Mitochondria and the pathogenesis of ALS.
 
Brain
, 123, 1291–2.

Beckman
JS, Carson M, Smith CD et al. (
1993
).
ALS, SOD and peroxynitrite.
 
Nature
, 364, 584.

Beetz
C, Nygren AOH, Schickel J et al. (
2006
).
High frequency of partial SPAST deletions in autosomal dominant hereditary spastic paraplegia.
 
Neurology
, 67, 1926–30.

Beetz
C, Zucnner S, Ashley-Koch A et al. (
2007
).
Linkage to a known gene but no mutation identified: comprehensive re-analysis of SPG4 HSP pedigrees reveals large deletions as the sole cause.
 
Human Mutation
, 28, 739–40.

Bence
NF, Sampat RM, Kopito RR (
2001
).
Impairment of the ubiquitin-proteasome system by protein aggregation.
 
Science
, 292, 1552–5.

Bendotti
C, Tortarolo M, Suchak SK et al. (
2001
).
Transgenic SOD1 G93A mice develop reduced GLT-1 in spinal cord without alterations in cerebrospinal fluid glutamate levels.
 
J Neurochem
, 79, 737–46.

Bensimon
G, Lacomblez L, Meininger V (
1994
).
A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group.
 
N Engl J Med
, 330, 585–91.

Blexrud
MD, Windebank AJ, Daube JR (
1993
).
Long-term follow-up of 121 patients with benign fasciculations.
 
Ann Neurol
, 34, 622–5.

Boillee
S, Yamanaka K, Lobsiger C. S. et al. (
2006
).
Onset and progression in inherited ALS determined by motor neurons and microglia.
 
Science
, 312, 1389–92.

Boltshauser
E, Lang W, Spillmann T et al (
1989
).
Hereditary distal muscular atrophy with vocal cord paralysis and sensorineural hearing loss: a dominant form of spinal muscular atrophy?
 
J Med Genet
, 26, 105–8.

Bonneau
D, Rozet JM, Bulteau C et al. (
1993
).
X linked spastic paraplegia (SPG2): clinical heterogeneity at a single gene locus.
 
J Med Genet
, 30, 381–4.

Borthwick
GM, Johnson MA, Ince PG et al. (
1999
).
Mitochondrial enzyme activity in amyotrophic lateral sclerosis: implications for the role of mitochondria in neuronal cell death.
 
Ann Neurol
, 46, 787–90.

Borthwick
GM, Taylor RW, Walls TJ et al. (
2006
).
Motor neuron disease in a patient with a mitochondrial tRNAIle mutation.
 
Ann Neurol
, 59, 570–4.

Bosch
EP, Yamada T, Kimura J (
1985
).
Somatosensory evoked potentials in motor neuron disease.
 
Muscle Nerve
, 8, 556–62.

Bouchard
JP, Barbeau A, Bouchard R et al. (
1978
).
Autosomal recessive spastic ataxia of Charlevoix-Saguenay.
 
Can J Neurol Sci
, 5, 61–9.

Bourke
SC, Bullock RE, Williams TL et al. (
2003
).
Noninvasive ventilation in ALS: indications and effect on quality of life.
 
Neurology
, 61, 171–7.

Bourke
SC, Tomlinson M, Williams TL et al. (
2006
).
Effects of non-invasive ventilation on survival and quality of life in patients with amyotrophic lateral sclerosis: a randomised controlled trial.
 
Lancet Neurol
, 5, 140–7.

Bourke
SC, Williams TL, Bullock RE et al. (
2002
).
Non-invasive ventilation in motor neuron disease: current UK practice.
 
Amyotroph Lateral Scler Other Motor Neuron Disord
, 3, 145–9.

Bowen
J, Gregory R, Squier M et al. (
1996
).
The post-irradiation lower motor neuron syndrome neuronopathy or radiculopathy?
 
Brain
, 119, 1429–39.

Brain
WR (
1933
).
Diseases of the Nervous System
. Oxford University Press, London, 1st edition, p. 801.

Brooks
BR (
1994
).
El Escorial World Federation of Neurology criteria for the diagnosis of amyotrophic lateral sclerosis.
Subcommittee on motor neuron diseases/amyotrophic lateral sclerosis of the World Federation of Neurology Research Group on neuromuscular diseases and the El Escorial “Clinical limits of amyotrophic lateral sclerosis” workshop contributors.
J Neurol Sci
, 124 (Suppl), 96–107.

Brooks
BR, Miller RG, Swash M, Munsat TL (
2000
).
El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis.
 
Amyotroph Lateral Scler Other Motor Neuron Disord
, 1, 293–9.

Brooks
BR, Shodis KA, Lewis DH et al. (
1995
).
Natural history of amyotrophic lateral sclerosis. Quantification of symptoms, signs, strength, and function.
 
Adv Neurol
, 68, 163–84.

Brownell
B, Oppenheimer DR, Hughes JT (
1970
).
The central nervous system in motor neurone disease.
 
J Neurol Neurosurg Psychiatry
, 33, 338–57.

Brucher
JM, Dom R, Lombaert A et al. (
1981
).
Progressive pontobulbar palsy with deafness: clinical and pathological study of two cases.
 
Arch Neurol
, 38, 186–90.

Brugman
F, Wokke JH, Scheffer H et al. (
2005
).
Spastin mutations in sporadic adult-onset upper motor neuron syndromes.
 
Ann Neurol
, 58, 865–9.

Bruijn
LI, Cleveland DW (
1996
).
Mechanisms of selective motor neuron death in ALS: insights from transgenic mouse models of motor neuron disease.
 
Neuropathol Appl Neurobiol
, 22, 373–87.

Bruijn
LI, Miller TM, Cleveland DW (
2004
).
Unraveling the mechanisms involved in motor neuron degeneration in ALS.
 
Annu Rev Neurosci
, 27, 723–49.

Bruyn
RP, Koelman JH, Troost D et al. (
1995
).
Motor neuron disease (amyotrophic lateral sclerosis) arising from longstanding primary lateral sclerosis.
 
J Neurol Neurosurg Psychiatry
, 58, 742–4.

Bruyn
RP, Scheltens P, Lycklama a Nijeholt J et al. (
1993
).
Autosomal recessive paraparesis with amyotrophy of the hands and feet.
 
Acta Neurol Scand
, 87, 443–5.

Bunina
TL (
1962
).
On intracellular inclusions in familial amyotrophic lateral sclerosis.
 
Zh Nevropatol Psikhiatr Im S S Korsakova
, 62, 1293–9.

Byrne
PC, Mc Monagle P, Webb S et al. (
2000
).
Age-related cognitive decline in hereditary spastic paraparesis linked to chromosome 2p.
 
Neurology
, 54, 1510–7.

Calza
L, Manfredi R, Freo E et al. (
2004
).
Transient reversal of HIV-associated motor neuron disease following the introduction of highly active antiretroviral therapy.
 
J Chemother
, 16, 98–101.

Caress
JB, Abend WK, Preston DC et al. (
1997
).
A case of Hodgkin’s lymphoma producing neuromyotonia.
 
Neurology
, 49, 258–9.

Carriedo
SG, Yin HZ, Weiss JH (
1996
). Motor neurons are selectively vulnerable to AMPA/kainate receptor-mediated injury in vitro.
J Neurosci
, 16, 4069–79.

Casali
C, Valente EM, Bertini E et al. (
2004
).
Clinical and genetic studies in hereditary spastic paraplegia with thin corpus callosum.
 
Neurology
, 62, 262–8.

Casari
G, De Fusco M, Ciarmatori S et al. (
1998
).
Spastic paraplegia and OXPHOS impairment caused by mutations in paraplegin, a nuclear-encoded mitochondrial metalloprotease.
 
Cell
, 93, 973–83.

Caselli
RJ, Smith BE, Osborne D (
1995
).
Primary lateral sclerosis: a neuropsychological study.
 
Neurology
, 45, 2005–9.

Castellani
V, Chedotal A, Schachner M et al. (
2000
).
Analysis of the L1-deficient mouse phenotype reveals cross-talk between Sema3A and L1 signaling pathways in axonal guidance.
 
Neuron
, 27, 237–49.

Chancellor
AM, Slattery JM, Fraser H et al. (
1993
).
The prognosis of adult-onset motor neuron disease: a prospective study based on the Scottish Motor Neuron Disease Register.
 
J Neurol
, 240, 339–46.

Chancellor
AM, Warlow CP (
1992
).
Adult onset motor neuron disease: worldwide mortality, incidence and distribution since 1950.
 
J Neurol Neurosurg Psychiatry
, 55, 1106–15.

Charcot
J-M, Joffroy A (
1869
).
Deux cas d’atrophie musculaire progressive avec lesions de la substance grise et de faisceaux anterolateraux de la moelle epiniere.
 
Arch Physiol Norm Pathol
, 3, 744–57.

Chen
YZ, Bennett CL, Huynh HM et al. (
2004
).
DNA/RNA helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis (ALS4).
 
Am J Hum Genet
, 74, 1128–35.

Chio
A, Benzi G, Dossena M et al. (
2005
).
Severely increased risk of amyotrophic lateral sclerosis among Italian professional football players.
 
Brain
, 128, 472–6.

Chotmongkol
V (
1999
).
Amyotrophic lateral sclerosis syndrome and hyperthyroidism: report of 4 patients.
 
J Med Assoc Thai
, 82, 615–8.

Christodoulou
K, Kyriakides T, Hristova AH et al. (
1995
).
Mapping of a distal form of spinal muscular atrophy with upper limb predominance to chromosome 7p.
 
Hum Mol Genet
, 4, 1629–32.

Ciccarelli
FD, Proukakis C, Patel H et al. (
2003
).
The identification of a conserved domain in both spartin and spastin, mutated in hereditary spastic paraplegia.
 
Genomics
, 81, 437–41.

Clark
LN, Poorkaj P, Wszolek Z et al. (
1998
).
Pathogenic implications of mutations in the tau gene in pallido-ponto-nigral degeneration and related neurodegenerative disorders linked to chromosome 17.
 
Proc Natl Acad Sci USA
, 95, 13103–7.

Clement
AM, Nguyen MD, Roberts EA et al. (
2003
).
Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice.
 
Science
, 302, 113–7.

Comi
GP, Bordoni A, Salani S et al. (
1998
).
Cytochrome c oxidase subunit I microdeletion in a patient with motor neuron disease.
 
Ann Neurol
, 43, 110–6.

Consortium.
Report (
1998
).
53rd ENMC International Workshop on Classification and Diagnostic Guidelines for Charcot-Marie-Tooth Type 2 and Distal Hereditary Motor Neuropathy
. Naarden, The Netherlands.

Cookson
MR, Menzies FM, Manning P et al. (
2002
).
Cu/Zn superoxide dismutase (SOD1) mutations associated with familial amyotrophic lateral sclerosis (ALS) affect cellular free radical release in the presence of oxidative stress.
 
Amyotroph Lateral Scler Other Motor Neuron Disord
, 3, 75–85.

Coovert
DD, Le TT, McAndrew PE et al. (
1997
).
The survival motor neuron protein in spinal muscular atrophy.
 
Hum Mol Genet
, 6, 1205–14.

Couillard-Despres
S, Zhu Q, Wong PC et al. (
1998
).
Protective effect of neurofilament heavy gene overexpression in motor neuron disease induced by mutant superoxide dismutase.
 
Proc Natl Acad Sci USA
, 95, 9626–30.

Cox
GA, Mahaffey CL, Frankel WN (
1998
).
Identification of the mouse neuromuscular degeneration gene and mapping of a second site suppressor allele.
 
Neuron
, 21, 1327–37.

Cox
PA, Banack SA, Murch SJ (
2003
).
Biomagnification of cyanobacterial neurotoxins and neurodegenerative disease among the Chamorro people of Guam.
 
Proc Natl Acad Sci USA
, 100, 13380–3.

Crosby
AH, Proukakis C (
2002
).
Is the transportation highway the right road for hereditary spastic paraplegia?
 
Am J Hum Genet
, 71, 1009–16.

Cross
HE, McKusick VA (
1967
a).
The mast syndrome. A recessively inherited form of presenile dementia with motor disturbances.
 
Arch Neurol
, 16, 1–13.

Cross
HE, McKusick VA (
1967
b).
The Troyer syndrome. A recessive form of spastic paraplegia with distal muscle wasting.
 
Arch Neurol
, 16, 473–85.

Crow
JP, Sampson JB, Zhuang Y et al. (
1997
).
Decreased zinc affinity of amyotrophic lateral sclerosis-associated superoxide dismutase mutants leads to enhanced catalysis of tyrosine nitration by peroxynitrite.
 
J Neurochem
, 69, 1936–44.

Dabby
R, Lange DJ, Trojaborg W et al. (
2001
).
Inclusion body myositis mimicking motor neuron disease.
Arch Neurol
 58, 1253–6.

Dahme
M, Bartsch U, Martini, R. et al. (
1997
).
Disruption of the mouse L1 gene leads to malformations of the nervous system.
 
Nat Genet
, 17, 346–9.

Dalakas
MC (
2006
).
Sporadic inclusion body myositis–diagnosis, pathogenesis and therapeutic strategies.
 
Nat Clin Pract Neurol
, 2, 437–47.

Davenport
RJ, Swingler RJ, Chancellor AM et al. (
1996
).
Avoiding false positive diagnoses of motor neuron disease: lessons from the Scottish Motor Neuron Disease Register.
 
J Neurol Neurosurg Psychiatry
, 60, 147–51.

de
Carvalho M, Matias T, Coelho F et al. (
1996
).
Motor neuron disease presenting with respiratory failure.
 
J Neurol Sci
, 139, 117–22.

de
Carvalho M, Swash M (
2004
).
Cramps, muscle pain, and fasciculations: not always benign?
 
Neurology
, 63, 721–3.

del
Aguila MA, Longstreth WT Jr, McGuire V et al. (
2003
).
Prognosis in amyotrophic lateral sclerosis: a population-based study.
 
Neurology
, 60, 813–9.

Demyanenko
GP, Tsai AY, Maness PF (
1999
).
Abnormalities in neuronal process extension, hippocampal development, and the ventricular system of L1 knockout mice.
 
J Neurosci
, 19, 4907–20.

Desport
JC, Preux PM, Truong TC et al. (
1999
).
Nutritional status is a prognostic factor for survival in ALS patients.
 
Neurology
, 53, 1059–63.

Dhaliwal
GK, Grewal RP (
2000
).
Mitochondrial DNA deletion mutation levels are elevated in ALS brains.
 
Neuroreport
, 11, 2507–9.

Dipti
S, Childs AM, Livingston JH et al. (
2005
).
Brown-Vialetto-Van Laere syndrome; variability in age at onset and disease progression highlighting the phenotypic overlap with Fazio-Londe disease.
 
Brain Dev
, 27, 443–6.

Donaghy
M, Kennett R (
1999
).
Varying occurrence of vocal cord paralysis in a family with autosomal dominant hereditary motor and sensory neuropathy.
 
J Neurol
, 246, 552–5.

Doyu
M, Sobue G, Mitsuma T et al. (
1993
).
Very late onset X-linked recessive bulbospinal neuronopathy: mild clinical features and a mild increase in the size of tandem CAG repeat in androgen receptor gene.
 
J Neurol Neurosurg Psychiatry
, 56, 832–3.

Doyu
M, Sobue G, Mukai E et al. (
1992
).
Severity of X-linked recessive bulbospinal neuronopathy correlates with size of the tandem CAG repeat in androgen receptor gene.
 
Ann Neurol
, 32, 707–10.

Drory
VE, Birnbaum M, Peleg L et al. (
2003
).
Hexosaminidase A deficiency is an uncommon cause of a syndrome mimicking amyotrophic lateral sclerosis.
 
Muscle Nerve
, 28, 109–12.

Durham
HD (
2003
).
Factors underlying the selective vulnerability of motor neurons to neurodegeneration.
 
Blue books of practical neurology: motor neuron disorders
.:379–400.

Edgar
JM, McLaughlin M, Yool D et al. (
2004
).
Oligodendroglial modulation of fast axonal transport in a mouse model of hereditary spastic paraplegia.
 
J Cell Biol
, 166, 121–31.

Erb
W (
1875
).
Uber einen wenig bekannten spinalen symptomenkomplex.
 
Berl Klin Wochenschr
, 12, 357–9.

Errico
A, Ballabio A, Rugarli EI (
2002
).
Spastin, the protein involved in autosomal dominant hereditary spastic paraparesis, is involved in microtubule dynamics.
 
Hum Mol Genet
, 11, 153–63.

Esteban
A, Traba A (
1993
).
Fasciculation-myokymic activity and prolonged nerve conduction block. A physiopathological relationship in radiation-induced brachial plexopathy.
 
Electroencephalogr Clin Neurophysiol
, 89, 382–91.

Estevez
AG, Crow JP, Sampson JB et al. (
1999
).
Induction of nitric oxide-dependent apoptosis in motor neurons by zinc-deficient superoxide dismutase.
 
Science
, 286, 2498–500.

Evans
BK, Fagan C, Arnold T (
1990
).
Paraneoplastic motor neuron disease and renal cell carcinoma: improvement after nephrectomy.
 
Neurology
, 40, 960–2.

Evgrafov
OV, Mersiyanova I, Irobi J et al. (
2004
).
Mutant small heat-shock protein 27 causes axonal Charcot-Marie-Tooth disease and distal hereditary motor neuropathy.
 
Nat Genet
, 36, 602–6.

Eymard-Pierre
E, Lesca G, Dollet S et al. (
2002
).
Infantile-onset ascending hereditary spastic paralysis is associated with mutations in the alsin gene.
 
Am J Hum Genet
, 71, 518–27.

Farbu
E, Gilhus NE, Barnes MP et al. (
2006
).
EFNS guideline on diagnosis and management of post-polio syndrome. Report of an EFNS task force.
 
Eur J Neurol
, 13, 795–801.

Feldkotter
M, Schwarzer V, Wirth R et al. (
2002
).
Quantitative analyses of SMN1 and SMN2 based on real-time lightCycler PCR: fast and highly reliable carrier testing and prediction of severity of spinal muscular atrophy.
 
Am J Hum Genet
, 70, 358–68.

Ferrante
MA, Wilbourn AJ (
1997
).
The characteristic electrodiagnostic features of Kennedy’s disease.
 
Muscle Nerve
, 20, 323–9.

Ferrante
RJ, Browne SE, Shinobu LA et al. (
1997
).
Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis.
 
J Neurochem
, 69, 2064–74.

Ferreirinha
F, Quattrini A, Pirozzi M et al. (
2004
).
Axonal degeneration in paraplegin-deficient mice is associated with abnormal mitochondria and impairment of axonal transport.
 
J Clin Invest
, 113, 231–42.

Ferrer
I, Olive M, Rivera R et al. (
1995
).
Hereditary spastic paraparesis with dementia, amyotrophy and peripheral neuropathy.
 
Neuropath Appl Neurobiol
, 21, 255–61.

Fetoni
V, Briem E, Carrara F et al. (
2004
).
Monomelic amyotrophy associated with the 7472insC mutation in the mtDNA tRNASer(UCN) gene.
 
Neuromuscul Disord
, 14, 723–6.

Fichera
M, Lo Giudice M, Flaco M et al. (
2004
).
Evidence of kinesin heavy chain (KIF5A) involvement in pure hereditary spastic paraplegia.
 
Neurology
, 63, 1108–10.

Figlewicz
DA, Krizus A, Martinoli MG et al. (
1994
).
Variants of the heavy neurofilament subunit are associated with the development of amyotrophic lateral sclerosis.
 
Hum Mol Genet
, 3, 1757–61.

Filiminoff
IN (
1926
).
Zur pathologisch-anatomischen Charakteristik des Lathyrismus.
 
Z Gesamte Neurol Psychiatr
, 105, 76–92.

Fischbeck
KH, Ionasescu V, Ritter AW et al. (
1986
).
Localization of the gene for X-linked spinal muscular atrophy.
 
Neurology
, 36, 1595–8.

Fisher
M, Mateer JE, Ullrich I, Gutrecht JA (
1985
).
Pyramidal tract deficits and polyneuropathy in hyperthyroidism, Combination clinically mimicking amyotrophic lateral sclerosis.
 
Am J Med
, 78, 1041–4.

Folli
F, Solimena M, Cofiell R et al. (
1993
).
Autoantibodies to a 128-kd synaptic protein in three women with the stiff-man syndrome and breast cancer.
 
N Engl J Med
, 328, 546–51.

Fonknechten
N, Mavel D, Byrne P et al. (
2000
).
Spectrum of SPG4 mutations in autosomal dominant spastic paraplegia.
 
Hum Mol Genet
, 9, 637–44.

Forbes
RB, Colville S, Swingler RJ (
2004
).
The epidemiology of amyotrophic lateral sclerosis (ALS/MND) in people aged 80 or over.
 
Age Ageing
, 33, 131–4.

Forsyth
PA, Dalmau J, Graus F et al. (
1997
).
Motor neuron syndromes in cancer patients.
 
Ann Neurol
, 41, 722–30.

Fotheringham
J, Jackson K, Kersh R, Gariballa SE (
2005
).
Refeeding syndrome: life-threatening, underdiagnosed, but treatable.
 
QJM
, 98, 318–9.

Fransen
E, Lemmon V, Van Camp G et al. (
1995
).
CRASH syndrome: clinical spectrum of corpus callosum hypoplasia, retardation, adducted thumbs, spastic paraparesis and hydrocephalus due to mutations in one single gene, L1.
 
Eur J Hum Genet
, 3, 273–84.

Fray
AE, Ince PG, Banner SJ et al. (
1998
).
The expression of the glial glutamate transporter protein EAAT2 in motor neuron disease: an immunohistochemical study.
 
Eur J Neurosci
, 10, 2481–9.

Freedman
DM, Travis LB, Gridley G et al. (
2005
).
Amyotrophic lateral sclerosis mortality in 1.9 million US cancer survivors.
 
Neuroepidemiology
, 25, 176–80.

Galasko
D, Salmon DP, Craig UK et al. (
2002
).
Clinical features and changing patterns of neurodegenerative disorders on Guam, 1997-2000.
 
Neurology
, 58, 90–7.

Gamez
J, Cervera C, Codina A. (
1999
).
Flail arm syndrome or Vulpian-Bernhart’s form of amyotrophic lateral sclerosis.
 
J Neurol Neurosurg Psychiatry
, 67, 258.

Gigli
GL, Diomedi M, Bernardi G et al. (
1993
).
Spastic paraplegia, epilepsy, and mental retardation in several members of a family: a novel genetic disorder.
 
Am J Med Genet
, 45, 711–6.

Glenn
SA, Ross MA (
2000
).
Delayed radiation-induced bulbar palsy mimicking ALS.
 
Muscle Nerve
, 23, 814–7.

Goldblatt
D (
1968
). Motor neuron disease: historical introduction. In
Motor Neuron Diseases
, Norris FH, Kurland LT (eds). New York: Grune and Stratton, 1968, pp. 3–11.

Gonzalez-Scarano
F, Baltuch G (
1999
).
Microglia as mediators of inflammatory and degenerative diseases.
 
Annu Rev Neurosci
, 22, 219–40.

Gonzalez
H, Sunnerhagen KS, Sjoberg I et al. (
2006
).
Intravenous immunoglobulin for post-polio syndrome: a randomised controlled trial.
 
Lancet Neurol
, 5, 493–500.

Gordon
PH, Rowland LP, Younger DS et al. (
1997
).
Lymphoproliferative disorders and motor neuron disease: an update.
 
Neurology
, 48, 1671–8.

Gourie-Devi
M, Nalini A (
2003
a).
Long-term follow-up of 44 patients with brachial monomelic amyotrophy.
 
Acta Neurol Scand
, 107, 215–20.

Gourie-Devi
M, Nalini A (
2003
b).
Madras motor neuron disease variant, clinical features of seven patients.
 
J Neurol Sci
, 209, 13–7.

Gourie-Devi
M, Suresh TG, Shankar SK (
1984
).
Monomelic amyotrophy.
 
Arch Neurol
, 41, 388–94.

Goytain
A, Hines R, El-Husseini A et al. (
2006
).
NIP1A (SPG6), the basis for autosomal dominant form of hereditary spastic paraplegia, encodes a functional Mg2+ transporter.
 
J Biol Chem
 (In Press).

Graus
F, Keime-Guibert F, Rene R et al. (
2001
).
Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients.
 
Brain
, 124, 1138–48.

Gregory
S, Siderowf A, Golaszewski AL et al. (
2002
).
Gastrostomy insertion in ALS patients with low vital capacity: respiratory support and survival.
 
Neurology
, 58, 485–7.

Griffin
JW, Li CY, Ho TW et al. (
1996
).
Pathology of the motor-sensory axonal Guillain-Barre syndrome.
 
Ann Neurol
, 39, 17–28.

Griffiths
I, Klugmann M, Anderson T et al. (
1998
a).
Current concepts of PLP and its role in the nervous system.
 
Microsc Res Tech
, 41, 344–58.

Griffiths
I, Klugmann M, Anderson T et al. (
1998
b).
Axonal swellings and degeneration in mice lacking the major proteolipid of myelin.
 
Science
, 280, 1610–3.

Griggs
RC, Askanas V, DiMauro S et al. (
1995
).
Inclusion body myositis and myopathies.
 
Ann Neurol
, 38, 705–13.

Grohmann
K, Schuelke M, Diers A et al. (
2001
).
Mutations in the gene encoding immunoglobulin mu-binding protein 2 cause spinal muscular atrophy with respiratory distress type 1.
 
Nat Genet
, 29, 75–7.

Gros-Louis
F, Meijer IA, Hand CK et al. (
2003
).
An ALS2 gene mutation causes hereditary spastic paraplegia in a Pakistani kindred.
 
Ann Neurol
, 53, 144–5.

Gudesblatt
M, Ludman MD, Cohen JA et al. (
1988
).
Hexosaminidase A activity and amyotrophic lateral sclerosis.
 
Muscle Nerve
, 11, 227–30.

Guegan
C, Przedborski S (
2003
).
Programmed cell death in amyotrophic lateral sclerosis.
 
J Clin Invest
, 111, 153–61.

Gurney
ME, Cutting FB, Zhai P et al. (
1996
).
Benefit of vitamin E, riluzole, and gabapentin in a transgenic model of familial amyotrophic lateral sclerosis.
 
Ann Neurol
, 39, 147–57.

Gutmann
L, Gutmann L (
2004
).
Myokymia and neuromyotonia 2004.
 
J Neurol
, 251, 138–42.

Hadano
S, Hand CK, Osuga H et al. (
2001
).
A gene encoding a putative GTPase regulator is mutated in familial amyotrophic lateral sclerosis 2.
 
Nat Genet
, 29, 166–73.

Hafezparast
M, Klocke R, Ruhrberg C et al. (
2003
).
Mutations in dynein link motor neuron degeneration to defects in retrograde transport.
 
Science
, 300, 808–12.

Hahn
AF, Parkes AW, Bolton CF et al. (
1991
).
Neuromyotonia in hereditary motor neuropathy.
 
J Neurol Neurosurg Psychiatry
, 54, 230–5.

Halperin
JJ, Kaplan GP, Brazinsky S et al. (
1990
).
Immunologic reactivity against Borrelia burgdorferi in patients with motor neuron disease.
 
Arch Neurol
, 47, 586–94.

Hansen
JJ, Durr A, Cournu-Rebeix I et al. (
2002
).
Hereditary spastic paraplegia SPG13 is associated with a mutation in the gene encoding the mitochondrial chaperonin Hsp60.
 
Am J Hum Genet
, 70, 1328–32.

Harding
AE (
1981
).
Hereditary “pure” spastic paraplegia: a clinical and genetic study of 22 families.
 
J Neurol Neurosurg Psychiatry
, 44, 871–83.

Harding
AE (
1993
).
Inherited neuronal atrophy and degeneration predominantly of lower motor neurones.
 
Peripheral Neuropathy
, 2, 1051–64.

Harding
AE, Thomas PK (
1984
).
Peroneal muscular atrophy with pyramidal features.
 
J Neurol Neurosurg Psychiatry
, 47, 168–72.

Hart
IK, Maddison P, Newsom-Davis J et al. (
2002
).
Phenotypic variants of autoimmune peripheral nerve hyperexcitability.
 
Brain
, 125, 1887–95.

Hart
IK, Waters C, Vincent A et al. (
1997
).
Autoantibodies detected to expressed K+ channels are implicated in neuromyotonia.
 
Ann Neurol
, 41, 238–46.

Haverkamp
LJ, Appel V, Appel SH (
1995
).
Natural history of amyotrophic lateral sclerosis in a database population. Validation of a scoring system and a model for survival prediction.
 
Brain
, 118, 707–19.

Hawkins
SA, Nevin NC, Harding AE (
1990
).
Pontobulbar palsy and neurosensory deafness (Brown-Vialetto-Van Laere syndrome) with possible autosomal dominant inheritance.
 
J Med Genet
, 27, 176–9.

Hayashi
H, Kato S (
1989
).
Total manifestations of amyotrophic lateral sclerosis. ALS in the totally locked-in state.
 
J Neurol Sci
, 93, 19–35.

Hazan
J, Fonknechten N, Mavel D et al. (
1999
).
Spastin, a new AAA protein, is altered in the most frequent form of autosomal dominant spastic paraplegia.
 
Nat Genet
, 23, 296–303.

Heath
P, Shaw PJ (
2005
). Excitotoxicity in amyotrophic lateral sclerosis. In
Amyotrophic Lateral Sclerosis
, H. Mitsumoto, S. Przedborski, P. Gordon, M. DelBene (eds). Marcel Dekker Inc, New York, pp. 297–336.

Hemmer
B, Glocker FX, Kaiser R et al. (
1997
).
Generalised motor neuron disease as an unusual manifestation of Borrelia burgdorferi infection.
 
J Neurol Neurosurg Psychiatry
, 63, 257–8.

Henningsen
P, Meinck HM (
2003
).
Specific phobia is a frequent non-motor feature in stiff man syndrome.
 
J Neurol Neurosurg Psychiatry
, 74, 462–5.

Hensley
K, Floyd RA, Gordon B et al. (
2002
).
Temporal patterns of cytokine and apoptosis-related gene expression in spinal cords of the G93A-SOD1 mouse model of amyotrophic lateral sclerosis.
 
J Neurochem
, 82, 365–74.

Hirano
A, Kurland LT, Krooth RS et al. (
1961
).
Parkinsonism-dementia complex, an endemic disease on the island of Guam. I. Clinical features.
 
Brain
, 84, 642–61.

Hirano
A, Kurland LT, Sayre GP (
1967
).
Familial amyotrophic lateral sclerosis. A subgroup characterized by posterior and spinocerebellar tract involvement and hyaline inclusions in the anterior horn cells.
 
Arch Neurol
, 16, 232–43.

Hirano
A, Llena JF, Streifler M et al. (
1976
).
Anterior horn cell changes in a case of neurolathyrism.
 
Acta Neuropathol
, 35, 277–83.

Hirayama
K, Tokokura Y, Tsubaki T (
1959
).
Juvenile muscular atrophy of unilateral upper extremity–a new clinical entity.
 
Psychiatry Neurology
, 61, 2190–97.

Hirayama
K, Tomonaga M, Kitano K et al. (
1987
).
Focal cervical poliopathy causing juvenile muscular atrophy of distal upper extremity: a pathological study.
 
J Neurol Neurosurg Psychiatry
, 50, 285–90.

Hirayama
K, Tsubaki T, Toyokura Y et al. (
1963
).
Juvenile muscular atrophy of unilateral upper extremity.
 
Neurology
, 13, 373–80.

Hirokawa
N (
1998
).
Kinesin and dynein superfamily proteins and the mechanism of organelle transport.
 
Science
, 279, 519–26.

Hodges
JR, Davies RR, Xuereb JH et al. (
2004
).
Clinicopathological correlates in frontotemporal dementia.
 
Ann Neurol
, 56, 399–406.

Horner
RD, Kamins KG, Feussner JR et al. (
2003
).
Occurrence of amyotrophic lateral sclerosis among Gulf War veterans.
 
Neurology
, 61, 742–9.

Hosler
BA, Siddique T, Sapp PC et al. (
2000
).
Linkage of familial amyotrophic lateral sclerosis with frontotemporal dementia to chromosome 9q21-q22.
 
JAMA
, 284, 1664–9.

Hsia
AW, Katz JS, Hancock SL, Peterson K (
2003
).
Post-irradiation polyradiculopathy mimics leptomeningeal tumor on MRI.
 
Neurology
, 60, 1694–6.

Hu
MT, Ellis CM, Al-Chalabi A et al. (
1998
).
Flail arm syndrome: a distinctive variant of amyotrophic lateral sclerosis.
 
J Neurol Neurosurg Psychiatry
, 65, 950–1.

Hudson
AJ (
1981
).
Amyotrophic lateral sclerosis and its association with dementia, parkinsonism and other neurological disorders: a review.
 
Brain
, 104, 217–47.

Hughes
CA, Byrne PC, Webb S et al. (
2001
).
SPG15, a new locus for autosomal recessive complicated HSP on chromosome 14q.
 
Neurology
, 56, 1230–3.

Hugon
J, Ludolph A, Gimenez-Roldan S et al. (
1990
). Electrophysiological evaluation of human lathyrism—results in Bangladesh and Spain. In
ALS New Advances in Toxicology and Epidemiology
, Clifford Rose F, Norris F (eds). Smith-Gordon, London, pp. 49–56.

Hurd
DD, Saxton WM (
1996
).
Kinesin mutations cause motor neuron disease phenotypes by disrupting fast axonal transport in Drosophila.
 
Genetics
, 144, 1075–85.

Ince
P. (
2000
).
Neuropathology.
 
Amyotrophic Lateral Sclerosis
, 83–12.

Ince
P, Stout N, Shaw P et al. (
1993
).
Parvalbumin and calbindin D-28k in the human motor system and in motor neuron disease.
 
Neuropathol Appl Neurobiol
, 19, 291–9.

Ince
PG, Codd GA (
2005
).
Return of the cycad hypothesis - does the amyotrophic lateral sclerosis/parkinsonism dementia complex (ALS/PDC) of Guam have new implications for global health?
 
Neuropathol Appl Neurobiol
, 31, 345–53.

Ince
PG, Evans J, Knopp M et al. (
2003
).
Corticospinal tract degeneration in the progressive muscular atrophy variant of ALS.
 
Neurology
, 60, 1252–8.

Ince
PG, Lowe J, Shaw PJ (
1998
a).
Amyotrophic lateral sclerosis: current issues in classification, pathogenesis and molecular pathology.
 
Neuropathol Appl Neurobiol
, 24, 104–17.

Ince
PG, Tomkins J, Slade JY et al. (
1998
b).
Amyotrophic lateral sclerosis associated with genetic abnormalities in the gene encoding Cu/Zn superoxide dismutase: molecular pathology of five new cases, and comparison with previous reports and 73 sporadic cases of ALS.
 
J Neuropathol Exp Neurol
, 57, 895–904.

Irobi
J, De Jonghe P, Timmerman V (
2004
a).
Molecular genetics of distal hereditary motor neuropathies.
 
Hum Mol Genet
, 13 Spec No 2, R195–202.

Irobi
J, Van den Bergh P, Merlini L et al. (
2004
b).
The phenotype of motor neuropathies associated with BSCL2 mutations is broader than Silver syndrome and distal HMN type V.
 
Brain
, 127, 2124–30.

Irobi
J, Van Impe K, Seeman P et al. (
2004
c).
Hot-spot residue in small heat-shock protein 22 causes distal motor neuropathy.
 
Nat Genet
, 36, 597–601.

Isozumi
K, DeLong R, Kaplan J et al. (
1996
).
Linkage of scapuloperoneal spinal muscular atrophy to chromosome 12q24.1-q24.31.
 
Hum Mol Genet
, 5, 1377–82.

Jackson
CE, Amato AA, Bryan WW et al. (
1998
).
Primary hyperparathyroidism and ALS: is there a relation?
 
Neurology
, 50, 1795–9.

Jaeckle
KA (
2004
).
Neurological manifestations of neoplastic and radiation-induced plexopathies.
 
Semin Neurol
, 24, 385–93.

Johnson
WG, Wigger HJ, Karp HR et al. (
1982
).
Juvenile spinal muscular atrophy: a new hexosaminidase deficiency phenotype.
 
Ann Neurol
, 11, 11–6.

Jokelainen
M (
1976
).
The epidemiology of amyotrophic lateral sclerosis in Finland. A study based on the death certificates of 421 patients.
 
J Neurol Sci
, 29, 55–63.

Kaback
M, Miles J, Jaffe M (
1978
).
Hexosaminidase A deficiency in early adulthood: A new type of GM2-gangliosidosis.
 
Am J Hum Genet
, 30, 30–31A.

Kaneko
K, Igarashi S, Miyatake T et al. (
1993
a).
‘Essential tremor’ and CAG repeats in the androgen receptor gene.
 
Neurology
, 43, 1618–9.

Kaneko
K, Igarashi S, Miyatake T et al. (
1993
b).
Hypertrophic cardiomyopathy and increased number of CAG repeats in the androgen receptor gene.
 
Am Heart J
, 126, 248–9.

Karni
A, Navon R, Sadeh M (
1988
).
Hexosaminidase A deficiency manifesting as spinal muscular atrophy of late onset.
 
Ann Neurol
, 24, 451–3.

Kato
S, Hayashi H, Yagishita A (
1993
).
Involvement of the frontotemporal lobe and limbic system in amyotrophic lateral sclerosis: as assessed by serial computed tomography and magnetic resonance imaging.
 
J Neurol Sci
, 116, 52–8.

Katsuno
M, Adachi H, Kume A et al. (
2002
).
Testosterone reduction prevents phenotypic expression in a transgenic mouse model of spinal and bulbar muscular atrophy.
 
Neuron
, 35, 843–54.

Katsuno
M, Adachi H, Tanaka F et al. (
2004
).
Spinal and bulbar muscular atrophy: ligand-dependent pathogenesis and therapeutic perspectives.
 
J Mol Med
, 82, 298–307.

Katz
JS, Wolfe GI, Andersson PB et al. (
1999
).
Brachial amyotrophic diplegia: a slowly progressive motor neuron disorder.
 
Neurology
, 53, 1071–6.

Kawamura
Y, Dyck PJ, Shimono M et al. (
1981
).
Morphometric comparison of the vulnerability of peripheral motor and sensory neurons in amyotrophic lateral sclerosis.
 
J Neuropathol Exp Neurol
, 40, 667–75.

Kawashima
T, Kikuchi H, Takita M et al. (
1998
).
Skein-like inclusions in the neostriatum from a case of amyotrophic lateral sclerosis with dementia.
 
Acta Neuropathol
, 96, 541–5.

Kelly
TE, Amoroso K, Ferre M et al. (
1999
).
Spinal muscular atrophy variant with congenital fractures.
 
Am J Med Genet
, 87, 65–8.

Kennedy
WR, Alter M, Sung JH (
1968
).
Progressive proximal spinal and bulbar muscular atrophy of late onset. A sex-linked recessive trait.
 
Neurology
, 18, 671–80.

Kertesz
A, McMonagle P, Blair M et al. (
2005
).
The evolution and pathology of frontotemporal dementia.
 
Brain
, 128, 1996–2005.

Khwaja
S, Sripathi N, Ahmad BK et al. (
1998
).
Paraneoplastic motor neuron disease with type 1 Purkinje cell antibodies.
 
Muscle Nerve
, 21, 943–5.

Kieran
D, Kalmar B, Dick JP et al. (
2004
).
Treatment with arimoclomol, a coinducer of heat shock proteins, delays disease progression in ALS mice.
 
Nat Med
, 10, 402–5.

Kirby
J, Heath PR, Allen S et al. (
2005
).
Mutant SOD1 alters the motor neurone transcriptome: implications for familial amyotrophic lateral sclerosis.
 
Brain
, 128, 1686–706.

Klivenyi
P, Ferrante RJ, Matthews RT et al. (
1999
).
Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis.
 
Nat Med
, 5, 347–50.

Kobayashi
H, Baumbach L, Matise TC et al. (
1995
).
A gene for a severe lethal form of X-linked arthrogryposis (X-linked infantile spinal muscular atrophy) maps to human chromosome Xp11.3-q11.2.
 
Hum Mol Genet
, 4, 1213–6.

Koerner
C, Wieland B, Richter W et al. (
2004
).
Stiff-person syndromes: motor cortex hyperexcitability correlates with anti-GAD autoimmunity.
 
Neurology
, 62, 1357–62.

Kogure
K, Busto R, Cassel J et al. (
1975
).
Effects of high-dose cyanate upon cerebral energy metabolism of the rat.
 
Pharmacology
, 13, 391–400.

Koul
R, Jain R, Chacko A et al. (
2006
).
Pontobulbar palsy and neurosensory deafness (Brown-Vialetto-van Laere syndrome) with hyperintense brainstem nuclei on magnetic resonance imaging: new finding in three siblings.
 
J Child Neurol
, 21, 523–5.

Kowalska
A, Konagaya M, Sakai M et al. (
2003
).
Familial amyotrophic lateral sclerosis and parkinsonism-dementia complex–tauopathy without mutations in the tau gene?
 
Folia Neuropathol
, 41, 59–64.

Kris
J, Millecamps S, Zhu Q (
2003
).
Creation of a mouse model for juvenile amuotrophic latgeral sclerosis.
 
Amyotroph Lateral Scler Other Motor Neuron Disord
, 4, 11.

Kruman
II, Pedersen WA, Springer JE et al. (
1999
).
ALS-linked Cu/Zn-SOD mutation increases vulnerability of motor neurons to excitotoxicity by a mechanism involving increased oxidative stress and perturbed calcium homeostasis.
 
Exp Neurol
, 160, 28–39.

Kuipers-Upmeijer
J, de Jager AE, Hew JM (
2001
).
Primary lateral sclerosis: clinical, neurophysiological, and magnetic resonance findings.
 
J Neurol Neurosurg Psychiatry
, 71, 615–20.

Kuncl
RW, Cornblath DR, Griffin JW (
1988
).
Assessment of thoracic paraspinal muscles in the diagnosis of ALS.
 
Muscle Nerve
, 11, 484–92.

Kunita
R, Otomo A, Mizumura H et al. (
2004
).
Homo-oligomerization of ALS2 through its unique carboxyl-terminal regions is essential for the ALS2-associated Rab5 guanine nucleotide exchange activity and its regulatory function on endosome trafficking.
 
J Biol Chem
, 279, 38626–35.

Kunst
CB (
2004
).
Complex genetics of amyotrophic lateral sclerosis.
 
Am J Hum Genet
, 75, 933–47.

Kurtzke
JF (
1991
).
Risk factors in amyotrophic lateral sclerosis.
 
Adv Neurol
, 56, 245–70.

Kusama-Eguchi
K, Kusama T, Suda A et al. (
2004
).
Partial involvement of group I metabotropic glutamate receptors in the neurotoxicity of 3-N-Oxalyl-L-2,3-diaminoprpanoic acid (L-beta-ODAP).
 
Biol Pharm Bull
, 27, 1052–8.

Kuzuhara
S, Kokubo Y (
2005
).
Atypical parkinsonism of Japan: amyotrophic lateral sclerosis-parkinsonism-dementia complex of the Kii peninsula of Japan (Muro disease): an update.
 
Mov Disord
, 20, S108–13.

La
Spada AR, Wilson EM et al. (
1991
).
Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy.
 
Nature
, 352, 77–9.

Lacomblez
L, Bensimon G, Leigh PN (
1996
).
Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis/Riluzole Study Group II.
 
Lancet
, 347, 1425–31.

Lagueny
A (
2005
).
Cramp-fasciculation syndrome.
 
Rev Neurol
, 161, 1260–6.

LaMonte
BH, Wallace KE, Holloway BA et al. (
2002
).
Disruption of dynein/dynactin inhibits axonal transport in motor neurons causing late-onset progressive degeneration.
 
Neuron
, 34, 715–27.

Lamy
C, Mas JL, Varet B, Ziegler M (
1991
).
Postradiation lower motor neuron syndrome presenting as monomelic amyotrophy.
 
J Neurol Neurosurg Psychiatry
, 54, 648–9.

Landau
D, Mishori-Dery A (
2003
).
A new autosomal recessive congenital contractural syndrome in an Israeli Bedouin kindred.
 
Am J Med Genet A
, 117, 37–40.

Langmore
SE, Kasarskis EJ, Manca ML et al. (
2006
).
Enteral tube feeding for amyotrophic lateral sclerosis/motor neuron disease.
 
Cochrane Database Syst Rev
, 4, CD004030.

Le
Forestier N, Maisonobe T, Piquard A et al. (
2001
).
Does primary lateral sclerosis exist? A study of 20 patients and a review of the literature.
 
Brain
, 124, 1989–99.

Lee
JH, Shin JH, Park KP et al. (
2005
).
Phenotypic variability in Kennedy’s disease: implication of the early diagnostic features.
 
Acta Neurol Scand
, 112, 57–63.

Lefebvre
S, Burlet P, Liu Q et al. (
1997
).
Correlation between severity and SMN protein level in spinal muscular atrophy.
 
Nat Genet
, 16, 265–9.

Leger
JM, Salachas F (
2001
).
Diagnosis of motor neuropathy.
 
Eur J Neurol
, 8, 201–8.

Leigh
PN, Anderton BH, Dodson A et al. (
1988
).
Ubiquitin deposits in anterior horn cells in motor neurone disease.
 
Neurosci Lett
, 93, 197–203.

Leigh
PN, Rothwell JC, Traub M et al. (
1980
).
A patient with reflex myoclonus and muscle rigidity: “jerking stiff-man syndrome
J Neurol Neurosurg Psychiatry
, 43, 1125–31.

Levy
LM, Levy-Reis I, Fujii M et al. (
2005
).
Brain gamma-aminobutyric acid changes in stiff-person syndrome.
 
Arch Neurol
, 62, 970–4.

Li
M, Miwa S, Kobayashi Y et al. (
1998
).
Nuclear inclusions of the androgen receptor protein in spinal and bulbar muscular atrophy.
 
Ann Neurol
, 44, 249–54.

Li
M, Ona VO, Chen M et al. (
2000
).
Functional role and therapeutic implications of neuronal caspase-1 and -3 in a mouse model of traumatic spinal cord injury.
 
Neuroscience
, 99, 333–42.

Lieberman
AP, Harmison G, Strand AD et al. (
2002
).
Altered transcriptional regulation in cells expressing the expanded polyglutamine androgen receptor.
 
Hum Mol Genet
, 11, 1967–76.

Lilienfeld
DE, Chan E, Ehland J et al. (
1989
).
Rising mortality from motoneuron disease in the USA, 1962-84.
 
Lancet
, 1, 710–3.

Lindsey
JC, Lusher ME, McDermott CJ et al. (
2000
).
Mutation analysis of the spastin gene (SPG4) in patients with hereditary spastic paraparesis.
 
J Med Genet
, 37, 759–65.

Liu
J, Lillo C, Jonsson PA et al. (
2004
).
Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria.
 
Neuron
, 43, 5–17.

Logina
I, Krumina A, Karelis G et al. (
2006
).
Clinical features of double infection with encephalitis (TBE) and Lyme Correliosis (LB) transmitted by tick bite.
 
J Neurol Neurosurg and Psychiatry
, 77, 1350–3.

Lorson
CL, Strasswimmer J, Yao, J. M. et al. (
1998
).
SMN oligomerization defect correlates with spinal muscular atrophy severity.
 
Nat Genet
, 19, 63–6.

Lowe
J, Aldridge F, Lennox G et al. (
1989
).
Inclusion bodies in motor cortex and brainstem of patients with motor neurone disease are detected by immunocytochemical localisation of ubiquitin.
 
Neurosci Lett
, 105, 7–13.

Lu
J, Rashid F, Byrne PC (
2006
).
The hereditary spastic paraplegia protein spartin localises to mitochondria.
 
J Neurochem
, 98, 1908–19.

Lyall
RA, Donaldson N, Fleming T et al. (
2001
).
A prospective study of quality of life in ALS patients treated with noninvasive ventilation.
 
Neurology
, 57, 153–6.

Maasilta
P, Jokelainen M, Loytonen M et al. (
2001
).
Mortality from amyotrophic lateral sclerosis in Finland, 1986-1995.
 
Acta Neurol Scand
, 104, 232–5.

Machida
Y, Tsuchiya K, Anno M et al. (
1999
).
Sporadic amyotrophic lateral sclerosis with multiple system degeneration: a report of an autopsy case without respirator administration.
 
Acta Neuropathol
, 98, 512–5.

Mackenzie
IR, Feldman H (
2003
).
The relationship between extramotor ubiquitin-immunoreactive neuronal inclusions and dementia in motor neuron disease.
 
Acta Neuropathol
, 105, 98–102.

Mackenzie
IR, Feldman HH (
2005
).
Ubiquitin immunohistochemistry suggests classic motor neuron disease, motor neuron disease with dementia, and frontotemporal dementia of the motor neuron disease type represent a clinicopathologic spectrum.
 
J Neuropathol Exp Neurol
, 64, 730–9.

Maddison
P (
2006
).
Neuromyotonia.
 
Clin Neurophysiol
, 117, 2118–27.

Maddison
P, Mills KR, Newsom-Davis J (
2006
).
Clinical electrophysiological characterization of the acquired neuromyotonia phenotype of autoimmune peripheral nerve hyperexcitability.
 
Muscle Nerve
, 33, 801–8.

Makela-Bengs
P, Jarvinen N, Vuopala K et al. (
1998
).
Assignment of the disease locus for lethal congenital contracture syndrome to a restricted region of chromosome 9q34, by genome scan using five affected individuals.
 
Am J Hum Genet
, 63, 506–16.

Mannan
AU, Krawen P, Sauter SM et al. (
2006
).
ZFYVE27 (SPG33), a novel spastin-binding protein, is mutated in hereditary spastic paraplegia.
 
Am J Hum Genet
, 79, 351–7.

Mannen
T, Iwata M, Toyokura Y et al. (
1977
).
Preservation of a certain motoneurone group of the sacral cord in amyotrophic lateral sclerosis: its clinical significance.
 
J Neurol Neurosurg Psychiatry
, 40, 464–9.

Martin
LJ (
1999
).
Neuronal death in amyotrophic lateral sclerosis is apoptosis: possible contribution of a programmed cell death mechanism.
 
J Neuropathol Exp Neurol
, 58, 459–71.

Matsuzaki
T, Nakagawa M, Nagai, M. et al. (
2000
).
HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP) with amyotrophic lateral sclerosis-like manifestations.
 
J Neurovirol
, 6, 544–8.

McCampbell
A, Taye AA, Whitty L et al. (
2001
).
Histone deacetylase inhibitors reduce polyglutamine toxicity.
 
Proc Natl Acad Sci USA
, 98, 15179–84.

McCampbell
A, Taylor JP, Taye AA et al. (
2000
).
CREB-binding protein sequestration by expanded polyglutamine.
 
Hum Mol Genet
, 9, 2197–202.

McDermott
CJ, Burness CE, Kirby J et al. (
2006
).
Clinical features of hereditary spastic paraplegia due to spastin mutation.
 
Neurology
, 67, 45–51.

McDermott
CJ, Dayaratne RK, Tomkins J et al. (
2001
).
Paraplegin gene analysis in hereditary spastic paraparesis (HSP) pedigrees in northeast England.
 
Neurology
, 56, 467–71.

McDermott
CJ, Grierson AJ, Wood JD et al. (
2003
a).
Hereditary spastic paraparesis: evidence of disrupted intracellular transport associated with spastin mutation.
 
Ann Neurol
, 54, 748–59.

McDermott
CJ, Taylor RW, Hayes C et al. (
2003
b).
Investigation of mitochondrial function in hereditary spastic paraparesis.
 
NeuroReport
, 14, 485–8.

McEntagart
M, Norton N, Williams H et al. (
2001
).
Localization of the gene for distal hereditary motor neuronopathy VII (dHMN-VII) to chromosome 2q14.
 
Am J Hum Genet
, 68, 1270–6.

McKhann
GM, Albert MS, Grossman M et al. (
2001
).
Clinical and pathological diagnosis of frontotemporal dementia: report of the Work Group on frontotemporal dementia and pick’s disease.
 
Arch Neurol
, 58, 1803–9.

McMonagle
P, Byrne, P, Hutchinson, M. (
2004
).
Further evidence of dementia in SPG4-linked autosomal dominant hereditary spastic paraplegia.
 
Neurology
, 62, 407–10.

McShane
MA, Boyd S, Harding B et al. (
1992
).
Progressive bulbar paralysis of childhood. A reappraisal of Fazio-Londe disease.
 
Brain
, 115, 1889–900.

McWhorter
ML, Monani UR, Burghes AH et al. (
2003
).
Knockdown of the survival motor neuron (Smn) protein in zebrafish causes defects in motor axon outgrowth and pathfinding.
 
J Cell Biol
, 162, 919–31.

Mead
SH, Proukakis C, Wood N et al. (
2001
).
A large family with hereditary spastic paraparesis due to a frame shift mutation of the spastin (SPG4) gene: association with multiple sclerosis in two affected siblings and epilepsy in other affected family members.
 
J Neurol Neurosurg Psychiatry
, 71, 788–91.

Meenakshisundaram
E, Jagannathan K, Ramamurthi B (
1970
).
Clinical pattern of motor neuron disease seen in younger age groups in Madras.
 
Neurol India
, 18 (Suppl 1), 109.

Meininger
V, Salachas F (
2000
).
Review of clinical trials.
 
Amyotroph Lateral Scler
: pp. 389–405.

Mellado
W, Slebe JC, Maccioni RB (
1982
).
Tubulin carbamoylation. Functional amino groups in microtubule assembly.
 
Biochem J
, 203, 675–81.

Menzies
FM, Cookson MR, Taylor RW et al. (
2002
a).
Mitochondrial dysfunction in a cell culture model of familial amyotrophic lateral sclerosis.
 
Brain
, 125, 1522–33.

Menzies
FM, Ince PG, Shaw PJ (
2002
b).
Mitochondrial involvement in amyotrophic lateral sclerosis.
 
Neurochem Int
, 40, 543–51.

Miller
RG, Mitchell JD, Lyon M et al. (
2002
).
Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND).
 
Cochrane Database Syst Rev
, 2, CD001447.

Miller
RG, Rosenberg JA, Gelinas DF et al. (
1999
). Practice parameter: the care of the patient with amyotrophic lateral sclerosis (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of
Neurology
: ALS Practice Parameters Task Force. Neurology, 52, 1311–23.

Mills
KR (
2003
).
Neurophysiological investigations of motor neuron disorders.
 
Blue Books of Practical Neurology: Motor Neuron Disorders
: pp 57–72.

Millul
A, Beghi E, Logroscino G et al. (
2005
).
Survival of patients with amyotrophic lateral sclerosis in a population-based registry.
 
Neuroepidemiology
, 25, 114–9.

Mitsumoto
H, Sliman RJ, Schafer IA et al. (
1985
).
Motor neuron disease and adult hexosaminidase A deficiency in two families: evidence for multisystem degeneration.
 
Ann Neurol
, 17, 378–85.

Mizutani
T, Aki M, Shiozawa R et al. (
1990
).
Development of ophthalmoplegia in amyotrophic lateral sclerosis during long-term use of respirators.
 
J Neurol Sci
, 99, 311–9.

Moersch
FP, Woltman HW (
1956
).
Progressive fluctuating muscular rigidity and spasm (“stiff-man” syndrome); report of a case and some observations in 13 other cases.
 
Mayo Clin Proc
, 31, 421–7.

Monani
UR, Sendtner M, Coovert DD et al. (
2000
).
The human centromeric survival motor neuron gene (SMN2) rescues embryonic lethality in Smn(-/-) mice and results in a mouse with spinal muscular atrophy.
 
Hum Mol Genet
, 9, 333–9.

Moreira
MC, Klur S, Watanabe M et al. (
2004
).
Senataxin, the ortholog of a yeast RNA helicase, is mutant in ataxia-ocular apraxia 2.
 
Nat Genet
, 36, 225–7.

Mostacciuolo
ML, Rampoldi L, Righetti E et al. (
2000
).
Hereditary spastic paraplegia associated with peripheral neuropathy: a distinct clinical and genetic entity.
 
Neuromuscul Disord
, 10, 497–502.

Mott
RT, Dickson DW, Trojanowski JQ et al. (
2005
).
Neuropathologic, biochemical, and molecular characterization of the frontotemporal dementias.
 
J Neuropathol Exp Neurol
, 64, 420–8.

Moulignier
A, Moulonguet A, Pialoux G et al. (
2001
).
Reversible ALS-like disorder in HIV infection.
 
Neurology
, 57, 995–1001.

Muglia
M, Magariello A, Nicoletti G et al. (
2002
).
Further evidence that SPG3A gene mutations cause autosomal dominant hereditary spastic paraplegia.
 
Ann Neurol
, 51, 794–5.

Munch
C, Rosenbohm A, Sperfeld AD et al. (
2005
).
Heterozygous R1101K mutation of the DCTN1 gene in a family with ALS and FTD.
 
Ann Neurol
, 58, 777–80.

Munch
C, Sedlmeier R, Meyer T et al. (
2004
).
Point mutations of the p150 subunit of dynactin (DCTN1) gene in ALS.
 
Neurology
, 63, 724–6.

Munoz
DG, Dickson DW, Bergeron C et al. (
2003
).
The neuropathology and biochemistry of frontotemporal dementia.
 
Ann Neurol
, 54, S24–8.

Munoz
DG, Greene C, Perl DP et al. (
1988
).
Accumulation of phosphorylated neurofilaments in anterior horn motoneurons of amyotrophic lateral sclerosis patients.
 
J Neuropathol Exp Neurol
, 47, 9–18.

Murakami
N (
1999
).
(Parkinsonism-dementia complex on Guam).
 
Ryoikibetsu Shokogun Shirizu
, (27 Pt 2), 57–60.

Mwanza
JC, Tshala-Katumbay D, Kayembe DL et al. (
2003
).
Neuro-ophthalmologic findings in konzo, an upper motor neuron disorder in Africa.
 
Eur J Ophthalmol
, 13, 383–9.

Nagai
M, Aoki M, Miyoshi I et al. (
2001
).
Rats expressing human cytosolic copper-zinc superoxide dismutase transgenes with amyotrophic lateral sclerosis: associated mutations develop motor neuron disease.
 
J Neurosci
, 21, 9246–54.

Nalini
A, Lokesh L, Ratnavalli E (
2004
).
Familial monomelic amyotrophy: a case report from India.
 
J Neurol Sci
, 220, 95–8.

Neary
D, Snowden JS, Gustafson L et al. (
1998
).
Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria.
 
Neurology
, 51, 1546–54.

Neary
D, Snowden JS, Mann DM et al. (
1990
).
Frontal lobe dementia and motor neuron disease.
 
J Neurol Neurosurg Psychiatry
, 53, 23–32.

Neumann
M, Sampathu DM, Kwong LK et al. (
2006
).
Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis.
 
Science
, 314, 130–3.

Nightingale
S, Bates D, Bateman DE et al. (
1982
).
Enigmatic dyspnoea: an unusual presentation of motor-neurone disease.
 
Lancet
, 1, 933–5.

Nishimura
AL, Mitne-Neto M, Silva HC et al. (
2004
).
A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis.
 
Am J Hum Genet
, 75, 822–31.

Ohara
S, Iwahashi T, Oide T et al. (
2002
).
Spinocerebellar ataxia type 6 with motor neuron loss: a follow-up autopsy report.
 
J Neurol
, 249, 633–5.

Okado-Matsumoto
A, Fridovich I (
2001
).
Subcellular distribution of superoxide dismutases (SOD) in rat liver: Cu,Zn-SOD in mitochondria.
 
J Biol Chem
, 276, 38388–93.

Okamoto
K, Hirai S, Amari M et al. (
1993
).
Bunina bodies in amyotrophic lateral sclerosis immunostained with rabbit anti-cystatin C serum.
 
Neurosci Lett
, 162, 125–8.

Olsen
NK, Pfeiffer P, Johannsen et al. (
1993
).
Radiation-induced brachial plexopathy: neurological follow-up in 161 recurrence-free breast cancer patients.
 
Int J Radiat Oncol Biol Phys
, 26, 43–9.

Ordway
JM, Tallaksen-Greene S, Gutekunst CA et al. (
1997
).
Ectopically expressed CAG repeats cause intranuclear inclusions and a progressive late onset neurological phenotype in the mouse.
 
Cell
, 91, 753–63.

Ostojic
J, Axelman K, Lannfelt L et al. (
2003
).
No evidence of linkage to chromosome 9q21-22 in a Swedish family with frontotemporal dementia and amyotrophic lateral sclerosis.
 
Neurosci Lett
, 340, 245–7.

Osuntokun
BO (
1981
).
Cassava diet, chronic cyanide intoxication and neuropathy in the Nigerian Africans.
 
World Rev Nutr Diet
, 36, 141–73.

Otomo
A, Hadano S, Okada, T. et al. (
2003
).
ALS2, a novel guanine nucleotide exchange factor for the small GTPase Rab5, is implicated in endosomal dynamics.
 
Hum Mol Genet
, 12, 1671–87.

Parboosingh
JS, Figlewicz DA, Krizus, A. et al. (
1997
).
Spinobulbar muscular atrophy can mimic ALS: the importance of genetic testing in male patients with atypical ALS.
 
Neurology
, 49, 568–72.

Pasinelli
P, Belford ME, Lennon, N. et al. (
2004
).
Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind and aggregate with Bcl-2 in spinal cord mitochondria.
 
Neuron
, 43, 19–30.

Pasinelli
P, Borchelt DR, Houseweart MK et al. (
1998
).
Caspase-1 is activated in neural cells and tissue with amyotrophic lateral sclerosis-associated mutations in copper-zinc superoxide dismutase.
 
Proc Natl Acad Sci USA
, 95, 15763–8.

Patel
H, Cross H, Proukakis C et al. (
2002
).
SPG20 is mutated in Troyer syndrome, an hereditary spastic paraplegia.
 
Nat Genet
, 31, 347–8.

Patel
H, Hart PE, Warner TT et al. (
2001
).
The Silver syndrome variant of hereditary spastic paraplegia maps to chromosome 11q12-q14, with evidence for genetic heterogeneity within this subtype.
 
Am J Hum Genet
, 69, 209–15.

Patel
S, Latterich, M. (
1998
).
The AAA team: related ATPases with diverse functions.
 
Trends Cell Biol
, 8, 65–71.

Patten
BM, Pages M (
1984
).
Severe neurological disease associated with hyperparathyroidism.
 
Ann Neurol
, 15, 453–6.

Peiris
JB, Seneviratne KN, Wickremasinghe HR et al. (
1989
).
Non familial juvenile distal spinal muscular atrophy of upper extremity.
 
J Neurol Neurosurg Psychiatry
, 52, 314–9.

Pellizzoni
L, Charroux B, Rappsilber J et al. (
2001
).
A functional interaction between the survival motor neuron complex and RNA polymerase II.
 
J Cell Biol
, 152, 75–85.

Pellizzoni
L, Kataoka N, Charroux B et al. (
1998
).
A novel function for SMN, the spinal muscular atrophy disease gene product, in pre-mRNA splicing.
 
Cell
, 95, 615–24.

Pelosi
L, Lanzillo B, Perretti A et al. (
1991
).
Motor and somatosensory evoked potentials in hereditary spastic paraplegia.
 
J Neurol Neurosurg Psychiatry
, 54, 1099–102.

Piao
YS, Wakabayashi K, Kakita A et al. (
2003
).
Neuropathology with clinical correlations of sporadic amyotrophic lateral sclerosis: 102 autopsy cases examined between 1962 and 2000.
 
Brain Pathol
, 13, 10–22.

Piccioni
F, Pinton P, Simeoni S et al. (
2002
).
Androgen receptor with elongated polyglutamine tract forms aggregates that alter axonal trafficking and mitochondrial distribution in motor neuronal processes.
 
Faseb J
, 16, 1418–20.

Plato
CC, Garruto RM, Galasko D et al. (
2003
).
Amyotrophic lateral sclerosis and parkinsonism-dementia complex of Guam: changing incidence rates during the past 60 years.
 
Am J Epidemiol
, 157, 149–57.

Prabhakar
S, Chopra JS, Banerjee AK et al. (
1981
).
Wasted leg syndrome: a clinical, electrophysiological and histopathological study.
 
Clin Neurol Neurosurg
, 83, 19–28.

Pridmore
S, Rao, G, Abusah, P. (
1995
).
Hereditary spastic paraplegia with dementia.
 
Aust N Z J Psychiatry
, 29, 678–82.

Pringle
CE, Hudson AJ, Munoz DG et al. (
1992
).
Primary lateral sclerosis. Clinical features, neuropathology and diagnostic criteria.
 
Brain
, 115, 495–520.

Proukakis
C, Cross H, Patel H et al. (
2004
).
Troyer syndrome revisited. A clinical and radiological study of a complicated hereditary spastic paraplegia.
 
J Neurol
, 251, 1105–10.

Puls
I, Jonnakuty C, LaMonte BH et al. (
2003
).
Mutant dynactin in motor neuron disease.
 
Nat Genet
, 33, 455–6.

Qureshi
AI, Wilmot G, Dihenia B et al. (
1996
).
Motor neuron disease with parkinsonism.
 
Arch Neurol
, 53, 987–91.

Ragonese
P, Fierro B, Salemi G et al. (
2005
).
Prevalence and risk factors of post-polio syndrome in a cohort of polio survivors.
 
J Neurol Sci
, 236, 31–5.

Rainier
S, Chai JH, Tokarz D et al. (
2003
).
NIPA1 gene mutations cause autosomal dominant hereditary spastic paraplegia (SPG6).
 
Am J Hum Genet
, 73, 967–71.

Raju
R, Rakocevic G, Chen Z et al. (
2006
).
Autoimmunity to GABAA-receptor-associated protein in stiff-person syndrome.
 
Brain
, 67, 1068–70.

Rakocevic
G, Raju, R, Dalakas MC (
2004
).
Anti-glutamic acid decarboxylase antibodies in the serum and cerebrospinal fluid of patients with stiff-person syndrome: correlation with clinical severity.
 
Arch Neurol
, 61, 902–4.

Rakocevic
G, Raju R, Semino-Mora C et al. (
2006
).
Stiff person syndrome with cerebellar disease and high-titer anti-GAD antibodies.
 
Neurology
, 67, 1068–70.

Ramlow
J, Alexander M, LaPorte R et al. (
1992
).
Epidemiology of the post-polio syndrome.
 
Am J Epidemiol
, 136, 769–86.

Rao
SD, Banack SA, Cox PA et al. (
2006
).
BMAA selectively injures motor neurons via AMPA/kainate receptor activation.
 
Exp Neurol
, 201, 244–52.

Ravindranath
V (
2002
).
Neurolathyrism: mitochondrial dysfunction in excitotoxicity mediated by L-beta-oxalyl aminoalanine.
 
Neurochem Int
, 40, 505–9.

Reid
E, Kloos M, Ashley-Koch A et al. (
2002
).
A kinesin heavy chain (KIF5A) mutation in hereditary spastic paraplegia (SPG10).
 
Am J Hum Genet
, 71, 1189–94.

Ringholz
GM, Appel SH, Bradshaw M et al. (
2005
).
Prevalence and patterns of cognitive impairment in sporadic ALS.
 
Neurology
, 65, 586–90.

Rodrigues
NR, Owen N, Talbot K et al. (
1996
).
Gene deletions in spinal muscular atrophy.
 
J Med Genet
, 33, 93–6.

Roeleveld-Versteegh
AB, Braun KP, Smeitink JA et al. (
2004
).
Mitochondrial respiratory chain disease presenting as progressive bulbar paralysis of childhood.
 
J Inherit Metab Dis
, 27, 281–3.

Rosati
G, Aiello I, Tola R et al. (
1980
).
Amyotrophic lateral sclerosis associated with thyrotoxicosis.
 
Arch Neurol
, 37, 530–1.

Rosen
DR, Siddique T, Patterson D et al. (
1993
).
Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis.
 
Nature
, 362, 59–62.

Rosenfeld
MR, Posner JB (
1991
).
Paraneoplastic motor neuron disease.
 
Adv Neurol
, 56, 445–59.

Rosling
H, Tylleskar T (
1995
).
Konzo.
 
Tropical Neurology
, 353–64.

Rosling
H, Tylleskar T (
2000
).
Cassava.
 
Exp Clin Neurotoxicol
, 338–43.

Rossoll
W, Jablonka S, Andreassi C et al. (
2003
).
Smn, the spinal muscular atrophy-determining gene product, modulates axon growth and localization of beta-actin mRNA in growth cones of motoneurons.
 
J Cell Biol
, 163, 801–12.

Rossoll
W, Kroning AK, Ohndorf UM et al. (
2002
).
Specific interaction of Smn, the spinal muscular atrophy determining gene product, with hnRNP-R and gry-rbp/hnRNP-Q: a role for Smn in RNA processing in motor axons?
 
Hum Mol Genet
, 11, 93–105.

Rothstein
JD, Van Kammen M, Levey AI et al. (
1995
).
Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis.
 
Ann Neurol
, 38, 73–84.

Rouleau
GA, Clark AW, Rooke K et al. (
1996
).
SOD1 mutation is associated with accumulation of neurofilaments in amyotrophic lateral sclerosis.
 
Ann Neurol
, 39, 128–31.

Rowland
LP (
1999
).
Primary lateral sclerosis: disease, syndrome, both or neither?
 
J Neurol Sci
, 170, 1–4.

Rowland
LP, Sherman WL, Hays AP et al. (
1995
).
Autopsy-proven amyotrophic lateral sclerosis, Waldenstrom’s macroglobulinemia, and antibodies to sulfated glucuronic acid paragloboside.
 
Neurology
, 45, 827–9.

Roy
N, Mahadevan MS, McLean M et al. (
1995
).
The gene for neuronal apoptosis inhibitory protein is partially deleted in individuals with spinal muscular atrophy.
 
Cell
, 80, 167–78.

Rudnik-Schoneborn
S, Forkert R, Hahnen E et al. (
1996
).
Clinical spectrum and diagnostic criteria of infantile spinal muscular atrophy: further delineation on the basis of SMN gene deletion findings.
 
Neuropediatrics
, 27, 8–15.

Rudnik-Schoneborn
S, Sztriha L, Aithala GR et al. (
2003
).
Extended phenotype of pontocerebellar hypoplasia with infantile spinal muscular atrophy.
 
Am J Med Genet A
, 117, 10–7.

Ryan
A, Nor AM, Costigan D et al. (
2003
).
Polymyositis masquerading as motor neuron disease.
 
Arch Neurol
, 60, 1001–3.

Sack
GH, Huether CA, Garg N (
1978
).
Familial spastic paraplegia-clinical and pathologic studies in a large kindred.
 
Johns Hopkins Med J
, 143, 117–21.

Saha
SP, Das SK, Gangopadhyay PK et al. (
1997
).
Pattern of motor neurone disease in eastern India.
 
Acta Neurol Scand
, 96, 14–21.

Sasaki
S, Tsutsumi Y, Yamane K et al. (
1992
).
Sporadic amyotrophic lateral sclerosis with extensive neurological involvement.
 
Acta Neuropathol
, 84, 211–5.

Sathasivam
S, Grierson AJ, Shaw PJ (
2005
).
Characterization of the caspase cascade in a cell culture model of SOD1-related familial amyotrophic lateral sclerosis: expression, activation and therapeutic effects of inhibition.
 
Neuropathol Appl Neurobiol
, 31, 467–85.

Sathasivam
S, O’Sullivan S, Nicolson A et al. (
2000
).
Brown-Vialetto-Van Laere syndrome: case report and literature review.
 
Amyotroph Lateral Scler Other Motor Neuron Disord
, 1, 277–81.

Sathasivam
S, Shaw PJ (
2005
).
Apoptosis in amyotrophic lateral sclerosis–what is the evidence?
 
Lancet Neurol
, 4, 500–9.

Schady
W, Dick JP, Sheard A Crampton S (
1991
).
Central motor conduction studies in hereditary spastic paraplegia.
 
J Neurol Neurosurg Psychiatry
, 54, 775–9.

Schady
W, Smith CM (
1994
).
Sensory neuropathy in hereditary spastic paraplegia.
 
J Neurol Neurosurg Psychiatry
, 57, 693–8.

Schreiber
H, Gaigalat T, Wiedemuth-Catrinescu U et al. (
2005
).
Cognitive function in bulbar- and spinal-onset amyotrophic lateral sclerosis. A longitudinal study in 52 patients.
 
J Neurol
, 252, 772–81.

Schroder
R, Keller E, Flacke S et al. (
1999
).
MRI findings in Hirayama’s disease: flexion-induced cervical myelopathy or intrinsic motor neuron disease?
 
J Neurol
, 246, 1069–74.

Seilhean
D, Takahashi J, El Hachimi KH et al. (
2004
).
Amyotrophic lateral sclerosis with neuronal intranuclear protein inclusions.
 
Acta Neuropathol
, 108, 81–7.

Seri
M, Cusano R, Forabosco P et al. (
1999
).
Genetic mapping to 10q23.3-q24.2, in a large Italian pedigree, of a new syndrome showing bilateral cataracts, gastroesophageal reflux, and spastic paraparesis with amyotrophy.
 
Am J Hum Genet
, 64, 586–93.

Shankar
SK, Gourie-Devi M, Shankar, L. et al. (
2000
).
Pathology of Madras type of motor neuron disease (MMND)–a histological and immunohistochemical study.
 
Acta Neuropathol
, 99, 428–34.

Shaw
PJ (
1999
).
Stiff-man syndrome and its variants.
 
Lancet
, 353, 86–7.

Shaw
PJ (
2005
).
Molecular and cellular pathways of neurodegeneration in motor neurone disease.
 
J Neurol Neurosurg Psychiatry
, 76, 1046–57.

Shaw
PJ, Bates D, Kendall-Taylor P (
1988
).
Hyperthyroidism presenting as pyramidal tract disease.
 
BMJ
, 297, 1395–6.

Shaw
PJ, Eggett CJ (
2000
).
Molecular factors underlying selective vulnerability of motor neurons to neurodegeneration in amyotrophic lateral sclerosis.
 
J Neurol
, 247, I17–27.

Shaw
PJ, Forrest V, Ince PG et al. (
1995
a).
CSF and plasma amino acid levels in motor neuron disease: elevation of CSF glutamate in a subset of patients.
 
Neurodegeneration
, 4, 209–16.

Shaw
PJ, Ince PG, Falkous G et al. (
1995
b).
Oxidative damage to protein in sporadic motor neuron disease spinal cord.
 
Ann Neurol
, 38, 691–5.

Shefner
JM, Tyler HR, Krarup C (
1991
).
Abnormalities in the sensory action potential in patients with amyotrophic lateral sclerosis.
 
Muscle Nerve
, 14, 1242–6.

Siklos
L, Engelhardt J, Harati Y et al. (
1996
).
Ultrastructural evidence for altered calcium in motor nerve terminals in amyotropic lateral sclerosis.
 
Ann Neurol
, 39, 203–16.

Silva
MT, Leite AC, Alamy AH et al. (
2005
).
ALS syndrome in HTLV-I infection.
 
Neurology
, 65, 1332–3.

Silver
JR (
1966
).
Familial spastic paraplegia with amyotrophy of the hands.
 
Ann Hum Genet
, 30, 69–75.

Simpson
MA, Cross H, Proukakis C et al. (
2003
).
Maspardin is mutated in mast syndrome, a complicated form of hereditary spastic paraplegia associated with dementia.
 
Am J Hum Genet
, 73, 1147–56.

Sinnreich
M, Klein CJ (
2002
).
Bulbospinal muscular atrophy: Kennedy’s disease.
 
Arch Neurol
, 61, 1324–6.

Slavotinek
AM, Pike M, Mills K et al. (
1996
).
Cataracts, motor system disorder, short stature, learning difficulties, and skeletal abnormalities: a new syndrome?
 
Am J Med Genet
, 62, 42–7.

Slee
M, Selvan A, Donaghy M (
2007
).
Multifocal motor neuropathy: the diagnostic spectrum and response to treatment.
 
Neurology
, 67, 1680–87.

Smith
RG, Henry YK, Mattson MP et al. (
1998
).
Presence of 4-hydroxynonenal in cerebrospinal fluid of patients with sporadic amyotrophic lateral sclerosis.
 
Ann Neurol
, 44, 696–9.

Sobue
G, Doyu M, Kachi T et al. (
1993
).
Subclinical phenotypic expressions in heterozygous females of X-linked recessive bulbospinal neuronopathy.
 
J Neurol Sci
, 117, 74–8.

Sobue
G, Hashizume Y, Yasuda T et al. (
1990
).
Phosphorylated high molecular weight neurofilament protein in lower motor neurons in amyotrophic lateral sclerosis and other neurodegenerative diseases involving ventral horn cells.
 
Acta Neuropathol
, 79, 402–8.

Sopher
BL, Thomas PS Jr, LaFevre-Bernt MA et al. (
2004
).
Androgen receptor YAC transgenic mice recapitulate SBMA motor neuronopathy and implicate VEGF164 in the motor neuron degeneration.
 
Neuron
, 41, 687–99.

Spalloni
A, Albo F, Ferrari F et al. (
2004
).
Cu/Zn-superoxide dismutase (GLY93–>ALA) mutation alters AMPA receptor subunit expression and function and potentiates kainate-mediated toxicity in motor neurons in culture.
 
Neurobiol Dis
, 15, 340–50.

Spencer
PS (
1999
).
Food toxins, ampa receptors, and motor neuron diseases.
 
Drug Metab Rev
, 31, 561–87.

Spencer
PS, Schaumburg HH, Cohn DF et al. (
1984
).
Lathyrism: a useful model of primary lateral sclerosis.
 
Research Progress in Motor Neuron Disease
, 312–27.

Spreux-Varoquaux
O, Bensimon G, Lacomblez L et al. (
2002
).
Glutamate levels in cerebrospinal fluid in amyotrophic lateral sclerosis: a reappraisal using a new HPLC method with coulometric detection in a large cohort of patients.
 
J Neurol Sci
, 193, 73–8.

Steffan
JS, Bodai L, Pallos J et al. (
2001
).
Histone deacetylase inhibitors arrest polyglutamine-dependent neurodegeneration in Drosophila.
 
Nature
, 413, 739–43.

Stolwijk-Swuste
JM, Beelen A, Lankhorst GJ et al. (
2005
).
The course of functional status and muscle strength in patients with late-onset sequelae of poliomyelitis: a systematic review.
 
Arch Phys Med Rehabil
, 86, 1693–701.

Striefler
M, Cohn DF, Hirano A et al. (
1977
).
The central nervous system in a case of neurolathyrism.
 
Neurology
, 27, 1176–8.

Strong
MJ, Gordon PH (
2005
).
Primary lateral sclerosis, hereditary spastic paraplegia and amyotrophic lateral sclerosis: discrete entities or spectrum?
 
Amyotroph Lateral Scler Other Motor Neuron Disord
, 6, 8–16.

Strong
MJ, Grace GM, Orange JB et al. (
1996
).
Cognition, language, and speech in amyotrophic lateral sclerosis: a review.
 
J Clin Exp Neuropsychol
, 18, 291–303.

Strong
MJ, Lomen-Hoerth C, Caselli RJ et al. (
2003
).
Cognitive impairment, frontotemporal dementia, and the motor neuron diseases.
 
Ann Neurol
, 54, S20–3.

Strumpell
A (
1886
).
Ueber eine bestimmte Form der primaren combinierten Systemerkrankung des Ruckenmarks.
 
Arch Psychiatr Nervenkr
, 17, 17–38.

Subramaniam
JR, Lyons WE, Liu J et al. (
2002
).
Mutant SOD1 causes motor neuron disease independent of copper chaperone-mediated copper loading.
 
Nat Neurosci
, 5, 301–7.

Sullivan
PG, Rabchevsky AG, Keller JN et al. (
2004
).
Intrinsic differences in brain and spinal cord mitochondria: Implication for therapeutic interventions.
 
J Comp Neurol
, 474, 524–34.

Takahashi
H, Snow BJ, Bhatt MH et al. (
1993
).
Evidence for a dopaminergic deficit in sporadic amyotrophic lateral sclerosis on positron emission scanning.
 
Lancet
, 342, 1016–8.

Takeuchi
H, Kobayashi Y, Ishigaki S et al. (
2002
).
Mitochondrial localization of mutant superoxide dismutase 1 triggers caspase-dependent cell death in a cellular model of familial amyotrophic lateral sclerosis.
 
J Biol Chem
, 277, 50966–72.

Taylor
BV, Gross L, Windebank AJ (
1996
).
The sensitivity and specificity of anti-GM1 antibody testing.
 
Neurology
, 47, 951–5.

Thompson
PD (
2001
).
The stiff-man syndrome and related disorders.
 
Parkinsonism Relat Disord
, 8, 147–53.

Thornton
FJ, Fotheringham T, Alexander M et al. (
2002
).
Amyotrophic lateral sclerosis: enteral nutrition provision–endoscopic or radiologic gastrostomy?
 
Radiology
, 224, 713–7.

Tikka
TM, Vartiainen NE, Goldsteins G et al. (
2002
).
Minocycline prevents neurotoxicity induced by cerebrospinal fluid from patients with motor neurone disease.
 
Brain
, 125, 722–31.

Tohgi
H, Abe T, Yamazaki K et al. (
1999
).
Remarkable increase in cerebrospinal fluid 3-nitrotyrosine in patients with sporadic amyotrophic lateral sclerosis.
 
Ann Neurol
, 46, 129–31.

Tomimitsu
H, Arimura K, Nagado, T. et al. (
2004
).
Mechanism of action of voltage-gated K+ channel antibodies in acquired neuromyotonia.
 
Ann Neurol
, 56, 440–4.

Tomkins
J, Usher PA, Slade JY et al. (
1998
).
Novel insertion in the KSP region of the neurofilament heavy gene in amyotrophic lateral sclerosis (ALS).
 
NeuroReport
, 9, 3967–70.

Traynor
BJ, Codd MB, Corr B et al. (
1999
).
Incidence and prevalence of ALS in Ireland, 1995-1997: a population-based study.
 
Neurology
, 52, 504–9.

Traynor
BJ, Codd MB, Corr, B. et al. (
2000
a).
Amyotrophic lateral sclerosis mimic syndromes: a population-based study.
 
Arch Neurol
, 57, 109–13.

Traynor
BJ, Codd MB, Corr B et al. (
2000
b).
Clinical features of amyotrophic lateral sclerosis according to the El Escorial and Airlie House diagnostic criteria: A population-based study.
 
Arch Neurol
, 57, 1171–6.

Trojan
DA, Cashman NR (
2005
).
Post-poliomyelitis syndrome.
 
Muscle Nerve
, 31, 6–19.

Troost
D, Van den Oord JJ, Vianney de Jong JM (
1990
).
Immunohistochemical characterization of the inflammatory infiltrate in amyotrophic lateral sclerosis.
 
Neuropathol Appl Neurobiol
, 16, 401–10.

Tshala-Katumbay
D, Eeg-Olofsson KE, Kazadi-Kayembe T et al. (
2002
).
Analysis of motor pathway involvement in konzo using transcranial electrical and magnetic stimulation.
 
Muscle Nerve
, 25, 230–5.

Tshala-Katumbay
D, Eeg-Olofsson KE, Tylleskar T et al. (
2001
).
Impairments, disabilities and handicap pattern in konzo–a non-progressive spastic para/tetraparesis of acute onset.
 
Disabil Rehabil
, 23, 731–6.

Turner
MR, Leigh PN (
2003
).
Disease modifying therapies in motor neurone disorders: the present position and potential future developments.
 
Motor Neuron Disorders
, 28, 497–545.

Turner
MR, Parton MJ, Shaw CE et al. (
2003
).
Prolonged survival in motor neuron disease: a descriptive study of the King’s database 1990-2002.
 
J Neurol Neurosurg Psychiatry
, 74, 995–7.

Tyler
HR, Sheffner J (
1991
).
Amyotrophic lateral sclerosis.
 
Handbook of Clinical Neurology
, 169–215.

Tylleskar
T, Howlett WP, Rwiza HT et al. (
1993
).
Konzo: a distinct disease entity with selective upper motor neuron damage.
 
J Neurol Neurosurg Psychiatry
, 56, 638–43.

Valdmanis
PN, Meijer IA, Reynolds A et al. (
2007
).
Mutations in the KIAA0196 gene at the SPG8 locus cause hereditary spastic paraplegia.
 
Am J Hum Genet
, 80, 152–61.

Van
Den Berg-Vos RM, Van Den Berg LH, Visser J et al. (
2003
a).
The spectrum of lower motor neuron syndromes.
 
J Neurol
, 250, 1279–92.

van
den Berg-Vos RM, Visser J, Franssen H et al. (
2003
b).
Sporadic lower motor neuron disease with adult onset: classification of subtypes.
 
Brain
, 126, 1036–47.

van
der Vleuten AJ, van Ravenswaaij-Arts CM, Frijns CJ et al. (
1998
).
Localisation of the gene for a dominant congenital spinal muscular atrophy predominantly affecting the lower limbs to chromosome 12q23-q24.
 
Eur J Hum Genet
, 6, 376–82.

Van
Es HW, Van den Berg LH, Franssen H et al. (
1997
).
Magnetic resonance imaging of the brachial plexus in patients with multifocal motor neuropathy.
 
Neurology
, 48, 1218–24.

Warner
TT, Patel H, Proukakis C et al. (
2004
).
A clinical, genetic and candidate gene study of Silver syndrome, a complicated form of hereditary spastic paraplegia.
 
J Neurol
, 251, 1068–74.

Watanabe
R, Iino M, Honda M (
1997
).
Primary lateral sclerosis.
 
Neuropathology
, 17, 220–4.

Webb
S, Coleman D, Byrne P et al. (
1998
).
Autosomal dominant hereditary spastic paraparesis with cognitive loss linked to chromosome 2p.
 
Brain
, 121, 601–9.

Weisskopf
MG, O’Reilly EJ, McCullough ML et al. (
2005
).
Prospective study of military service and mortality from ALS.
 
Neurology
, 64, 32–7.

Wharton
SB, McDermott CJ, Grierson AJ et al. (
2003
).
The cellular and molecular pathology of the motor system in hereditary spastic paraparesis due to mutation of the spastin gene.
 
J Neuropathol Exp Neurol
, 62, 1166–77.

White
KD, Ince PG, Lusher M et al. (
2000
).
Clinical and pathologic findings in hereditary spastic paraparesis with spastin mutation.
 
Neurology
, 55, 89–94.

Whiteley
AM, Swash M, Urich H (
1976
).
Progressive encephalomyelitis with rigidity.
 
Brain
, 99, 27–42.

Wiedemann
FR, Manfredi G, Mawrin C et al. (
2002
).
Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients.
 
J Neurochem
, 80, 616–25.

Wilhelmsen
KC, Blake DM, Lynch T et al. (
1996
).
Chromosome 12-linked autosomal dominant scapuloperoneal muscular dystrophy.
 
Ann Neurol
, 39, 507–20.

Willeit
J, Kiechl S, Kiechl-Kohlendorfer U et al. (
2001
).
Juvenile asymmetric segmental spinal muscular atrophy (Hirayama’s disease): three cases without evidence of “flexion myelopathy”.
 
Acta Neurol Scand
, 104, 320–2.

Williams
TL, Day NC, Ince PG et al. (
1997
).
Calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors: a molecular determinant of selective vulnerability in amyotrophic lateral sclerosis.
 
Ann Neurol
, 42, 200–7.

Williamson
TL, Cleveland DW (
1999
).
Slowing of axonal transport is a very early event in the toxicity of ALS-linked SOD1 mutants to motor neurons.
 
Nat Neurosci
, 2, 50–6.

Wilson
CM, Grace GM, Munoz DG et al. (
2001
).
Cognitive impairment in sporadic ALS: a pathologic continuum underlying a multisystem disorder.
 
Neurology
, 57, 651–7.

Windpassinger
C, Auer-Grumbach M, Irobi J et al. (
2004
).
Heterozygous missense mutations in BSCL2 are associated with distal hereditary motor neuropathy and Silver syndrome.
 
Nat Genet
, 36, 271–6.

Wirth
B. (
2000
).
An update on the mutation spectrum of the survival motor neuron gene (SMN1) in autosomal recessive spinal muscular atrophy (SMA).
 
Hum Mutat
, 15, 228–37.

Wong
PC, Pardo CA, Borchelt DR et al. (
1995
).
An adverse property of a familial ALS-linked SOD1 mutation causes motor neuron disease characterized by vacuolar degeneration of mitochondria.
 
Neuron
, 14, 1105–16.

Wood-Allum
CA, Barber SC, Kirby J et al. (
2006
).
Impairment of mitochondrial anti-oxidant defence in SOD1-related motor neuron injury and amelerioration by ebselen.
 
Brain
, 129, 1693–709.

Yamanaka
K, Vande Velde C, Eymard-Pierre E et al. (
2003
).
Unstable mutants in the peripheral endosomal membrane component ALS2 cause early-onset motor neuron disease.
 
Proc Natl Acad Sci USA
, 100, 16041–6.

Yang
Y, Hentati A, Deng HX et al. (
2001
).
The gene encoding alsin, a protein with three guanine-nucleotide exchange factor domains, is mutated in a form of recessive amyotrophic lateral sclerosis.
 
Nat Genet
, 29, 160–5.

Yih
JS, Wang SJ, Su MS et al. (
1993
).
Hereditary spastic paraplegia associated with epilepsy, mental retardation and hearing impairment.
 
Paraplegia
, 31, 408–11.

Yong
J, Wan L, Dreyfuss G (
2004
).
Why do cells need an assembly machine for RNA-protein complexes?
 
Trends Cell Biol
, 14, 226–32.

Younger
DS, Rowland LP, Latov N et al. (
1990
).
Motor neuron disease and amyotrophic lateral sclerosis: relation of high CSF protein content to paraproteinemia and clinical syndromes.
 
Neurology
, 40, 595–9.

Zeitlman
L, Sirim P, Kremmer E, Kolanus W (
2001
).
Cloning of ACP33 as a novel intracellular ligand of CD4.
 
J Biol Chem
, 276, 9123–32.

Zerres
K, Rudnik-Schoneborn S (
1995
).
Natural history in proximal spinal muscular atrophy. Clinical analysis of 445 patients and suggestions for a modification of existing classifications.
 
Arch Neurol
, 52, 518–23.

Zhao
Q, Wang J, Levichkin IV et al. (
2002
).
A mitochondrial specific stress response in mammalian cells.
 
Embo J
, 21, 4411–9.

Zhao
X, Alvarado D, Rainier S et al. (
2001
).
Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia.
 
Nat Genet
, 29, 326–31.

Zhu
PP, Patterson A, Lavoie B et al. (
2003
).
Cellular localization, oligomerization, and membrane association of the hereditary spastic paraplegia 3A (SPG3A) protein atlastin.
 
J Biol Chem
, 278, 49063–71.

Zhu
S, Stavrovskaya IG, Drozda M et al. (
2002
).
Minocycline inhibits cytochrome c release and delays progression of amyotrophic lateral sclerosis in mice.
 
Nature
, 417, 74–8.

Zoghbi
HY, Orr HT (
2000
).
Glutamine repeats and neurodegeneration.
 
Annu Rev Neurosci
, 23, 217–47.

Zuchner
S, Wang G, Tran-Viet K-N et al. (
2006
).
Mutations in the novel mitochondrial protein REEP1 cause hereditary spastic paraplegia type 31.
 
Am J Hum Genet
, 79, 365–9.

Close
This Feature Is Available To Subscribers Only

Sign In or Create an Account

Close

This PDF is available to Subscribers Only

View Article Abstract & Purchase Options

For full access to this pdf, sign in to an existing account, or purchase an annual subscription.

Close