Skip to main content
Review Article
Originally Published 4 August 2017
Free Access

Brain–Heart Interaction: Cardiac Complications After Stroke

Abstract

Neurocardiology is an emerging specialty that addresses the interaction between the brain and the heart, that is, the effects of cardiac injury on the brain and the effects of brain injury on the heart. This review article focuses on cardiac dysfunction in the setting of stroke such as ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage. The majority of post-stroke deaths are attributed to neurological damage, and cardiovascular complications are the second leading cause of post-stroke mortality. Accumulating clinical and experimental evidence suggests a causal relationship between brain damage and heart dysfunction. Thus, it is important to determine whether cardiac dysfunction is triggered by stroke, is an unrelated complication, or is the underlying cause of stroke. Stroke-induced cardiac damage may lead to fatality or potentially lifelong cardiac problems (such as heart failure), or to mild and recoverable damage such as neurogenic stress cardiomyopathy and Takotsubo cardiomyopathy. The role of location and lateralization of brain lesions after stroke in brain–heart interaction; clinical biomarkers and manifestations of cardiac complications; and underlying mechanisms of brain–heart interaction after stroke, such as the hypothalamic–pituitary–adrenal axis; catecholamine surge; sympathetic and parasympathetic regulation; microvesicles; microRNAs; gut microbiome, immunoresponse, and systemic inflammation, are discussed.
Cardiac injury is common in patients with cerebrovascular disease.13 In 1947, Byer et al4 first reported that cerebral vascular disease can cause myocardial damage and arrhythmia. The specialty that deals with the brain–heart connection is now referred to as neurocardiology.5 Typically, stroke (ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage [SAH]) induces neurovascular uncoupling and disrupts cerebral autoregulation, which then renders cerebral blood flow directly dependent on cardiac function.6 Multiple interactions occur among the various forms of cardiovascular and cerebrovascular diseases. Myocardial injury, ischemia-like electrocardiographic changes, and arrhythmias are frequently encountered in acute stroke patients, even in the absence of primary heart disease, which support a central nervous system (CNS) origin of these electrocardiographic abnormalities.13,7 Using a meta-analysis, including 25 studies with a total of 2690 patients, van der Bilt et al8 found that cardiac dysfunction is associated with an increased risk of death, delayed cerebral ischemia, and poor outcome after SAH. On the basis of accumulating clinical evidence, it is likely that there exists a causal relationship between brain damage and heart dysfunction. It is important to determine whether cardiac dysfunction in patients is triggered by stroke, is an unrelated complication, or is the underlying cause of stroke. Therefore, investigating the brain–heart interaction after stroke is highly clinically significant.

Scope of This Review

A multitude of brain injuries such as stroke (ischemic stroke, brain hemorrhage, or SAH), traumatic brain injury (TBI), brain tumor, and various causes of intracranial hypertension can lead to cardiac dysfunction, arrhythmias, and heart failure. Brain–heart syndrome broadly refers to heart damage caused by various brain disorders. The ability to accurately diagnose the occurrence and development of brain–heart syndrome is highly valued in the clinic. This review article focuses on brain–heart interaction after stroke of CNS origin. The role of location and lateralization of brain lesions in brain–heart interaction, clinical manifestations, pathophysiology, and mechanisms of brain–heart interaction after stroke are discussed.

Cardiac Damage and Stroke in the Light of Risk Factors and Pre-Existing Heart Disease

Cardiac complications leading to morbidity and even mortality in the days immediately after an acute stroke include heart attack, congestive heart failure, cardiac arrest, and abnormal heart rhythms like atrial fibrillation.5 Converging risk factors for cerebrovascular and cardiovascular diseases, such as hypertension, diabetes mellitus, high cholesterol, and age, exacerbate cardiac injury irrespective of stroke cause or subtype.9 Therefore, severe heart problems are more likely caused by systemic dysfunction–induced vascular damage, inflammation, and immune responses, such as in hypertension and diabetes mellitus, rather than a direct neural causation, although brain damage may act to worsen cardiac dysfunction.10,11 The Framingham study reported that stroke incidence more than doubled in the presence of coronary heart disease, more than tripled with hypertension, increased 4-fold with cardiac failure, and increased 5-fold with atrial fibrillation.9 Approximately 20% of ischemic strokes are caused by cardiac disease, the major risk factor being atrial fibrillation.9 In addition to being the most common tachyarrhythmia in acute stroke, atrial fibrillation is also highly associated with an increased risk of systemic thromboembolism.9,12 A history of cardiovascular diseases and hypertension increases incidence of electrocardiographic abnormalities in comparison to ischemic stroke patients with no primary heart disease.13 The aging population is largely affected by heart failure because of structural or functional damage to the heart affecting ventricular blood filling and ejection fraction.14 In this review, to specifically elucidate the brain–heart interaction of CNS origin in the setting of stroke, we discuss the mechanisms of brain–heart interaction after stroke without pre-existing heart disease. How diabetes mellitus, hypertension, age, sex, atrial fibrillation, and pre-existing heart disease influence brain–heart interaction are beyond the scope of this review and are not discussed.

Cardiac Damage as a Consequence of Stroke

The second leading cause of post-stroke death is cardiovascular complications.15 Broadly, stroke-induced cardiac damage may lead to fatality, potentially lifelong cardiac problems (such as heart failure), or mild and recoverable damage, such as neurogenic stress cardiomyopathy (NSC) and Takotsubo cardiomyopathy. Cardiac dysfunction manifested during the acute phase after brain injury usually resolves over the following several weeks alongside improvement of neurological function.15 Stabilization of the patient in relation to the type of stroke takes precedence over treatment of the heart dysfunction. NSC is diagnosed by reduced left ventricular (LV) ejection fraction, ventricular wall motion abnormalities, and elevated serum cardiac enzymes. Although Takotsubo cardiomyopathy shares the symptoms and transient nature of NSC, it is caused by psychological stress in the absence of physical damage to the brain. The telltale sign of Takotsubo cardiomyopathy is apical ballooning, caused by a weakening of the heart’s muscular cells. Still, Takotsubo cardiomyopathy and NSC may induce ventricular wall motion abnormalities in other regions of the heart.16,17 Many studies focus on the impaired contraction of the ventricle, which results in low ejection fraction, but it seems that the relaxation of the ventricle is also disrupted in NSC. Clinical symptoms of cardiac dysfunction after brain hemorrhagic stroke and ischemic stroke are summarized in Table 1.
Table 1. Summary of Clinical Manifestation of Cardiac Dysfunction After Stroke in Patients
 SAHISComments
TTCTTC is a transient left ventricular apical ballooning syndromeWomen are more prone to TTC and risk increases with age (IS in post-menopausal stage)15TTC is often misdiagnosed as acute coronary heart disease172
It is typically triggered by physical or emotional stressPatients are at risk for recurrence
It is diagnosed by elevated cardiac enzymes, WMA, and ECG changes
Stress-induced cardiomyopathyDyspnea and chest pain, ECG abnormalities such as ST-segment elevation, T-wave inversion, prolonged QT interval, and echocardiographic WMA172Patients may also experience post-stroke depression
TTC-like LV dysfunction and delayed cerebral ischemia have been reported172Early ECG abnormalities (few hours to 48 h after IS onset)15
Infarcts in/near insular cortex15
Transient LV EF reduction15
Abnormal cardiac function and ECG abnormalitiesReduced ventricular EFHigh troponin levels are related to the severity of reduced EF and LVDD173
Cardiac dysfunction increases with severity of stroke or hemorrhage
75% stroke patients have some ECG abnormality11
Cardiac arrhythmias during acute phase (≈37.5% SAH patients)18Cardiac arrhythmias (≈22% IS patients) and LVDD (≈50% IS patients)24Acute IS in right cerebral hemisphere increases susceptibility and severity of arrhythmias1
WMA (15% to 22% SAH patients)8LVDD may or may not be symptomatic but both cases hinder functional recovery18,19
Hypokinetic WMASevere LVDD (≈10% IS patients) may indicate stroke with atrial fibrillation175
ECG ventricular repolarization disturbances such as ST-segment elevation and T-wave inversion that mimics acute myocardial infarction174Impaired cardiac function after acute IS may predict a 3-mo mortality176Patients have greater ECG abnormalities and are susceptible to developing supraventricular tachycardia177
Secondary complicationsDiastolic dysfunction correlated with pulmonary edema11,176Vasospasm with increased risk of delayed cerebral ischemia172Severity of diastolic dysfunction increases composite vascular events at 1 y24
Recovery of cardiac functionCorrected QT prolongation, bradycardia recovers immediately after aneurysmal clipping21LVH can persist at 7 d21LVDD persists in TTC when systolic function recovers at predischarge180
cTnI levels return to normal by day 7178ST-segment elevation/depression, T-wave inversion persists at 7 d in some patients21TTC after stroke may be reversible24
RWMA abnormalities tend to be temporary and improves in 66% patients at follow-up179
cTn indicates cardiac troponin; EF, ejection fraction; IS, ischemic stroke; LV, left ventricular; LVDD, left ventricular diastolic dysfunction; LVEF, left ventricular ejection fraction; LVH, left ventricular hypertrophy; RWMA, regional wall motion abnormalities; SAH, subarachnoid hemorrhage; TTC, Takotsubo cardiomyopathy; and WMA, wall motion abnormalities.

Hemorrhagic Stroke–Induced Cardiac Damage

Experimental animal studies and clinical data indicate a gradient of NSC, where electrocardiographic abnormalities are found in 40% to 100% of SAH patients and 5% SAH patients have serious cardiac arrhythmias.18 Cardiac arrhythmias are associated with an increased risk of cardiovascular comorbidity, prolonged hospital stay, and poor outcome or death after SAH.18 The life-threatening ventricular arrhythmia is also associated with elevated creatine phosphokinase myocardial fraction mass and increased troponin T level in left side of intracerebral hemorrhagic stroke patients.19 Eighty percent of patients exhibit ischemic-like electrocardiographic changes within 1 year after intracerebral hemorrhage or SAH.20 In SAH patients, the Hunt–Hess grading score of severity shows that electrocardiographic changes are observed consistently at the lower grades, but echocardiography changes are found predominately in patients with higher grades.21 It is likely that less severe electrocardiographic changes precede structural alterations, as QTc (the interval between peak contraction and repolarization) prolongation does not predict ventricular wall motion abnormalities, but more severe electrocardiographic changes like inverted T waves and elevated Troponin levels, secreted by damaged sarcomeres, can predict ventricular wall motion abnormalities.16 Lee et al22 found that in spontaneous and traumatic brain hemorrhage patients, 7.2% (no history of cardiac disease patients) show acute cardiac dysfunction and 43% have LV hypertrophy.22 Acute cardiac dysfunction is independently associated with in-hospital death22,23 and associated with a diminished 1-year functional outcome.20 LV diastolic dysfunction reflects abnormalities in atrial filling by venous return, atrial contraction, and ventricular relaxation capabilities. Although LV diastolic dysfunction is a cause of cardioembolic stroke (present in 15%–25% of the population, increasing with age and hypertension24), more than half of SAH patients develop LV diastolic dysfunction, despite experiencing a stroke of noncardioembolic origin.

Ischemic Stroke–Induced Cardiac Damage

The risk of cardiac complications increases proportionally to the severity of ischemic stroke and neurological deficits.15 Likewise, impaired cardiac function as a consequence of severe acute ischemic stroke is a predictor of worse functional outcome and secondary complications.25 Clinically relevant secondary complications such as vasospasm, delayed cerebral ischemia, and pulmonary edema require active management. Recent evidence suggests that ischemic stroke can cause cardiac dysfunction even in the absence of risk factors and pre-existing heart disease.26 During the first 3 months after acute ischemic stroke, 19.0% of patients experience at least 1 serious cardiac adverse event; 28.5% have LV ejection fraction <50%; and 13% to 29% have systolic dysfunction.27,28 Approximately 67% of acute ischemic stroke patients have electrocardiographic abnormalities of ischemic and arrhythmic in the first 24 hours after stroke.29 Cardiac arrhythmias are common reasons for death after acute ischemic stroke.26 About 88% of stroke patients who sustain damage to the insular cortex in the right cerebral hemisphere develop myocardial injury in the weeks after ischemic stroke.1
Stroke (lacunar subtype) also induces heart problems in ≤70% of patients, with clinical manifestations such as electrocardiographic changes, reduced LV ejection fraction, ventricular wall motion abnormalities, and increases in serum cardiac enzymes.28,30 However, there is a paucity of studies investigating the mechanisms of small or lacunar stroke-induced cardiac dysfunction in both clinical and experimental research.

Myocardial Enzymes

cTnI (cardiac troponin I) is considered a more specific and sensitive biomarker for the detection of cardiac damage and LV dysfunction than CK-MB.31 CK-MB is not completely cardiac specific and may increase in skeletal muscle injury, kidney failure, intramuscular injection, strenuous exercise, and after exposure to several toxins and drugs.31 CK-MB elevations are most likely of noncardiac origin in patients with large hemispheric stroke.31 Increased circulating cTnI is associated with poor clinical outcome and cardiac function, such as ST-segment elevation in myocardial infarction patients,32 as well as in patients with advanced myocardial hypertrophy, fibrosis, and cardiovascular death.33,34
Serum cTnI or cTnT (cardiac troponin T) levels are indicators of cardiac damage after ischemic stroke, intracerebral hemorrhage stroke, and SAH. Elevation of serum myocardial enzymes is reported in 11% to 21% of SAH patients and in 1% to 17% of ischemic stroke patients.35 SAH patients have higher levels of circulating hsTnT (high-sensitivity troponin T) and NT-proBNP (N-terminal fragment of B-type natriuretic peptide).36,37 Twenty-two percent of brain hemorrhage patients have TnI elevation and 59% have NT-proBNP elevation.38 In acute ischemic stroke patients, 5% to 8% patients have high level of circulating cTnI and cTnT,39,40 and 65% patients have increased levels of circulating NT-proBNP.40 A recent study reported that patients who had acute ischemic stroke in the dorsal anterior insular cortical region of the right hemisphere had elevated relative changes of high-sensitivity cTnT levels, which can induce autonomic dysbalance, sympathetic activation, and myocardial injury.41
The level of hsTnT and NT-proBNP are strongly related with poor long-term outcome in SAH patients and are associated with increased stroke severity, mortality, and worse neurological functional outcome after ischemic stroke.36,42,43 However, using multivariate regression analysis, Etgen et al44 reported that neither cardiac troponins nor NT-proBNP influence morbidity and mortality if other risk factors are considered. Nigro et al42 found that BNP, but not hsTnT level, predicts short- and long-term prognosis after cerebrovascular events. Among the reported independent predictors of elevated cTnI are female sex, larger body surface area, and higher heart rate.45 The American Stroke Association recommends a baseline electrocardiographic and baseline troponin assessment in patients with acute stroke to identify concurrent myocardial ischemia or cardiac arrhythmias.46 Myocardial enzymes and biomarkers of cardiac dysfunction after brain hemorrhage and ischemic stroke are summarized in Table 2.
Table 2. Summary of Biomarkers of Cardiac Dysfunction After Stroke in Patients
 SAHISComments
CK-MBCK-MB level is associated with poor outcome181CK-MB elevation after stroke may not be of cardiac origin31CK-MB is not completely cardiac specific and may increase in skeletal muscle injury, kidney failure, intramuscular injection, strenuous exercise, and after exposure to several toxins and drugs31
Modest and progressive increase in expression
Elevated for a few days
cTn(cTnT and cTnI)cTnI level peaks within 24–36 h after SAH37Elevated cTns are observed in 5%–8% of IS patients39,40cTnT is a specific and sensitive biomarker for cardiac damage
High cTnT levels correlated with LV hypokinesia182Circulating cTnI level is an independent predictor of stroke prognosis39The American Stroke Association recommends a routine examination of cardiac troponin in patients with acute stroke
Elevated cTnI levels is associated with increased risk of LVDD, in-hospital morbidity, and mortality45,178,183Serum cTnT expression helps to evaluate myocardial injury and estimate ischemic lesion volume and is closely associated with the risk of death in patients with acute IS and severe neurological deficits at stroke onset and damage to the insular cortex41
May indicate delayed cerebral ischemia from vasospasmMay indicate coincidence of acute coronary syndrome or coronary artery disease43
CRPElevated CRP level is an independent predictor of future stroke and transient ischemic attack184CRP is a plasma protein that increases in response to inflammation during the acute phase of brain injury186
Patients with an increased plasma CRP level within 72 h of stroke onset battle high mortality because of cardiovascular complications185CRP may serve as a clinically useful risk marker for the development of unstable atherosclerotic disease, as well as a predictor of future cardiovascular morbidity and mortality186
Stroke patients with CRP levels >1.5 mg/dL (optimum sensitivity and specificity for adverse outcome) have a worse prognosis185
Patients with elevated CRP levels beyond the inflammatory acute phase and at time of hospital discharge face a danger of subsequent cardiovascular complications, death, and severe neurological deficit and disability, which should be carefully factored in their follow-up treatment185
NT-proBNPNT-proBNP increases the risk of death and delayed cerebral ischemia40NT-proBNP levels is increased after ischemic stroke40NT-proBNP is the product of pro-BNP (brain natriuretic peptide precursor) being cleaved into BNP and is mainly found in myocardial cells
Increased NT-proBNP levels is an independent risk factor for stroke40NT-proBNP is synthesized and secreted by normal cardiac muscle cells, and levels can rapidly increase after myocardial injury or necrosis
Increased NT-proBNP levels is associated with unfavorable functional outcome and mortality rates after stroke43
NT-proBNP is an indicator of ischemic stroke of cardioembolic cause
About 10-fold increase in NT-proBNP when TTC develops after stroke15
CK-MB indicates creatine kinase-MB; CRP, C-reactive protein; cTn, cardiac troponin; IS, ischemic stroke; LV, left ventricular; LVDD, left ventricular diastolic dysfunction; NT-proBNP, N-terminal probrain natriuretic peptide; SAH, subarachnoid hemorrhage; and TTC, Takotsubo cardiomyopathy.

Heart Rate Variability

Heart rate variability (HRV) serves as an assessment tool for autonomic cardiac control and is a measurement of the variation in the heart beat interval (interval between successive R peaks in the QRS complex of ECG) reflecting the instantaneous fluctuation in heart rate. Heart rate fluctuation may result from the neurohumoral regulation of the body to adapt to different physiological conditions, or in response to certain pathological conditions. Heart rhythm oscillations may be categorized into 4 primary frequency bands: high-frequency (HF), low-frequency (LF), very-low-frequency, and ultralow-frequency. Respiration modulates vagal (parasympathetic) activity that contributes to the HF component of heart rate variability.47 LF is thought to be the combined sympathetic and vagal response to arterial baroreceptors.48 Although not an accurate measure, it is commonly interpreted that a low LF to HF ratio reflects greater parasympathetic activity relative to sympathetic activity, whereas a high LF to HF ratio indicates higher sympathetic activity relative to parasympathetic activity.47 Dysfunction in the CNS can have downstream effects on spinal cord preganglionic autonomic nerves and result in HRV changes. Changes in HRV may represent an inadequate response of the autonomic nervous system to the stress associated with a more severe condition.49 Studies have indicated a neural source of HRV changes in ischemic stroke patients.50 In SAH patients, lowered LF/HF ratio is decreased in patients with pulmonary edema and indicates poor quality recovery and high mortality.51 Left hemisphere lesions in ischemic stroke patients induce increased HF (parasympathetic) rhythms, whereas right hemisphere lesions increase the LF/HF ratio.52 HRV decrease is associated with acute stroke severity, post-stroke depression, poor-quality recovery, and mortality.49,53

Mechanisms of Brain–Heart Interaction After Stroke

The mechanisms of brain–heart interaction resulting in cardiac dysfunction after injury to the brain are discussed below and summarized in Figure 1.
Figure 1. Summary of mechanisms for brain–heart interaction after stroke. Cardiac dysfunction after stroke may be caused by several mechanisms, including activation of the hypothalamic–pituitary–adrenal (HPA) axis, sympathetic and parasympathetic regulation, catecholamine surge, gut microbiome dysbiosis, immune responses, and inflammation, as well as the release of microvesicles and microRNA (? indicates that future studies are warranted).

Hypothalamic–Pituitary–Adrenal Axis, Catecholamine Surge, and Sympathetic and Parasympathetic Regulation After Stroke

Many patients develop early-onset depression related to the degree of their functional deficits or late-onset depression.54,55 Post-stroke depression including anxiety, fear, and stress is psychological and biochemical in nature and is largely dependent on the extent of neurological functional deficits.54,55 Ischemic stress can cause Takotsubo cardiomyopathy.15 The HPA axis is a master regulator of body hormones that integrates emotion, stress, physical activity state, and metabolism.56 The HPA axis consisting of the complex interactions between the 3 endocrine glands—hypothalamus, pituitary, and adrenal glands—is an important part of the neuroendocrine system. The hypothalamic paraventricular nucleus is the main control center of the HPA and secretes corticotropin-releasing factor such as corticotrophin-releasing hormone and vasopressin that stimulate the pituitary gland to release adrenocorticotropic hormone, especially under stressful conditions.57 Adrenocorticotropic hormone stimulates the adrenal gland to release the steroid hormone cortisol. Serum cortisol levels have been correlated with stroke severity and extent of insular damage.58 Prolonged elevation of cortisol may be neurotoxic and has been associated with increased post-stroke mortality.59 The paraventricular nucleus also projects directly to the rostral ventrolateral medulla, which integrates cardiac afferents, baroreceptor activity, and inputs from higher brain regions into sympathetic outflow to the heart. Stimulation of the hypothalamus activates sympathetic output and induces electrocardiographic abnormalities, arrhythmias, and myocardial necrosis.60 Activation of the paraventricular nucleus of the hypothalamus in rats subjected to ischemic stroke causes arrhythmias mediated by glutamate via activation of N-methyl-d-aspartic acid receptors.61 Reduction of paraventricular nucleus activity promotes improved recovery of cardiac function after myocardial infarction.62 Activation of the HPA axis after ischemic stroke causes a significant increase in catecholamines.63 The catecholamine surge hypothesis is the most widely accepted mechanism of brain–heart interaction.
The catecholamine surge theory has been closely linked to heart damage after physical and emotional stressors. Catecholamine surge can cause cardiac hypertrophy or myocardial ischemia.64,65 The autonomic nervous system regulates the release of catecholamines from the adrenal glands. Brain injury can cause elevated sympathetic tone with an additional increase in catecholamine secretion.66 Neurological injury causes excessive circulating catecholamines and massive catecholamine release from myocardial nerve endings.66 The myocardium adjacent to the nerve is damaged.67 The sympathetic nerve can directly release catecholamines and thereby induce cardiomyocyte toxicity. Long-term elevation of serum catecholamine produces cardiotoxicity68 and may trigger edema in regions of hypokinesia, transient fibrosis, inflammation, and contraction band necrosis.69,70 Experimental animal studies have demonstrated an increase in plasma catecholamine levels after ischemic stroke that was directly proportional to the incidence of myocardial lesions and cardiac damage.71 The increased catecholamine levels is correlated with QT-interval prolongation and myocardial damage after SAH, whereas hypothalamic stimulation induces electrocardiographic changes without associated myocardial damage.72,73
Catecholamines act on the heart via β receptors to increase the strength and rate of contraction.74 The β receptors couple to the stimulatory G protein and activate adenylyl cyclase and increase cytosolic cAMP. cAMP binds to the regulatory subunits of protein kinase A, which phosphorylates sarcolemmal L-type Ca2+ channels and sarcoplasmic phospholamban, which overloads mitochondria.75 Mitochondrial Ca2+ overload triggers oxidative stress, and the subsequent opening of their inner membrane permeability transition pore along with osmotic swelling and loss of ATP synthesis leads to myocardial cell death.75 Although pretreatment with β-blockers has been reported to decrease the incidence of myocardial lesion76 and β blockers are commonly prescribed after Takotsubo cardiomyopathy, some studies have reported a lack of significant effect of β blockers on recurrence of Takotsubo syndrome rate.77
Sympathetic and parasympathetic physiology and regulation of brain–heart interaction has been described in detail elsewhere.78 The forebrain plays an important role in the regulation of the autonomic nervous system in patients with hemorrhagic and ischemic stroke. Stimulating the orbital surface of the frontal lobe and the anterior cingulate gyrus affects blood pressure and heart rate.79 The insular cortex is regarded as a vital part of the central autonomic network.80 Ischemic lesions in the insular cortex increase the risk of cardiac complication and can cause blood pressure variations, cardiac arrhythmias, and myocytolysis.81,82 The location of an ischemic lesion also affects cardiac function after stroke, and studies have reported cardiac dysfunction after ischemic damage to both left and right insular cortex.52,80,83 It seems that the right hemisphere primarily controls sympathetic activity, whereas parasympathetic activity is mainly controlled by the left hemisphere.71,72 Therefore, a right insular lesion decreases sympathetic tone and results in parasympathetic overactivity.80 Right insular lesions are associated with higher mortality at an early stage compared with other sites.84 Brain infarctions in the left hemisphere are associated with fewer incidences of arrhythmias, an increased risk of adverse cardiac outcome, increased long-term mortality, and decreased cardiac wall motion compared with stroke in other locations.85,86 Elevated activity of the sympathetic nervous system observed in the acute phase of SAH induces myocardial damage and contributes to the development of cardiac dysfunction.87 Therefore, intensive heart monitoring will be particularly essential for patients with insular cortical damage. Figure 2 summarizes the role of the HPA axis, catecholamine surge, and sympathetic regulation in mediating cardiac dysfunction after stroke.
Figure 2. The hypothalamic–pituitary–adrenal (HPA) axis, catecholamine surge, and sympathetic and parasympathetic regulation mediate cardiac dysfunction after stroke. Catecholamines act on β receptors, β receptors couple to the stimulatory G protein (Gs) and activate adenylyl cyclase (AC), and increase cytosolic cAMP. cAMP binds to the regulatory subunits of protein kinase A (PKA), which phosphorylates sarcolemmal L-type Ca2+ channels (LTCC), and sarcoplasmic phospholamban, which overloads mitochondria. Mitochondrial Ca2+ overload triggers oxidative stress and the subsequent opening of their inner membrane permeability transition pore with ensuing osmotic swelling, and loss of ATP synthesis leads to myocardial cell death.

Blood–Brain Barrier Disruption After Stroke

The neurovascular unit is a functional unit encompassing the anatomic and metabolic interactions between the neuronal, glial, and vascular components of the brain. The blood–brain barrier (BBB) composed of endothelial cells, astrocytic end feet, pericytes, and a thick basement membrane is at the core of the neurovascular unit.88,89 The BBB serves as a dynamic interface between the brain and the circulatory system, limiting the entry of potential neurotoxins and microscopic and hydrophilic molecules.88,89 The BBB also protects the brain from variations in blood composition and helps maintain constant and optimal cerebral metabolism and neuronal activity. Disruption of the neurovascular unit and BBB is a major step in the pathophysiological cascade after stroke and contributes to secondary tissue damage. Increased BBB permeability disturbs cerebral autoregulation, leads to neuronal damage, and facilitates the invasion of inflammatory factors into the injured brain.90 In a vicious cycle, BBB disruption promotes entry of inflammatory factors into the ischemic brain, and inflammation increases BBB permeability. Increased BBB permeability is found in almost half the patients at 24 hours post-cardiac surgery even in the absence of stroke.91 Additionally, cardiac dysfunction and cardiac surgery can lead to cerebral ischemia and stroke via hypoperfusion and cardiac embolization, respectively, and trigger systemic inflammatory responses that may either induce or aggravate BBB disruption.91 BBB disruption facilitates the entry of brain-derived antigens and extracellular vesicles derived from injured brain cells to enter the blood stream and encounter peripheral immune cells, as described in detail in the following section. Therefore, there exists a close relationship between cardiac function and BBB integrity, but the effects are most likely indirect.

Immunoresponse and Systemic Inflammation After Stroke

Immune system activation leading to inflammation after stroke is an important factor in stroke progression. The complex interplay of the local and systemic inflammatory responses after stroke involves many immune cell types and circulating signals and is important in understanding both infection and other secondary complications. The complex role of inflammation in stroke pathogenesis has been reviewed elsewhere.92,93 In the following section, we discuss inflammatory and immune responses that may mediate brain–heart interaction after stroke, which is summarized in Figure 3.
Figure 3. Role of immune response and inflammation in mediating cardiac dysfunction after stroke. Local inflammatory responses in the brain, endothelial cell damage, and increased oxidative stress lead to blood–brain barrier (BBB) disruption after stroke. Release of proinflammatory cytokine and chemokines can be stimulated by damaged endothelial cells and astrocytes, activated resident microglia, and infiltrating macrophages that assume M1 phenotype, as well as the spleen. Systemic inflammatory responses are mainly driven by many cytokines, chemokines, stress hormones, as well as parasympathetic and sympathetic regulation. Gut microbiome dysbiosis after stroke can also lead to bacterial and endotoxin translocation to the blood, leading to systemic inflammation. Damage-associated molecular patterns released by dying brain cells after ischemic stroke release brain-derived antigens that can pass the ruptured BBB and enter the systemic circulation. In the presence of systemic inflammation, the antigen-specific autoimmune response leads to the secretion of proinflammatory cytokines. GFAP indicates glial fibrillary acidic protein; ICAM, intercellular adhesion molecule; IL, interleukin; MBP, myelin basic protein; MV, microvesicle; ROS, reactive oxygen species; TNF, tumor necrosis factor; and VCAM, vascular cell adhesion protein.
In the acute phase of brain injury, local inflammatory responses in the brain parenchyma include microgliosis, astrogliosis, and cytokine/chemokine secretion, which occur concomitantly with endothelial cell activation.94 Ischemia can lead to endothelial cell damage directly or indirectly via the release of reactive oxygen species.95 Damaged endothelial cells can increase oxidative stress and rupture the BBB.95 In addition, damaged endothelial cells and astrocytes at the site of inflammation can shed extracellular vesicles that rapidly cross the BBB and enter the bloodstream.96 On reaching peripheral organs such as the liver, the protein and microRNA (miR) cargo of these endothelial cell and astrocyte-derived extracellular vesicles regulate acute cytokine response and mediate trafficking of peripheral immune cells to injured brain tissue.96 Extracellular vesicles are emerging as intrinsic communication mediators between the brain and immune system.96 The role of microvesicles and miR as mediators of brain damage induced heart dysfunction is discussed in detail later in this review.
BBB disruption facilitates the infiltration of peripheral macrophages and neutrophils into brain after stroke onset.97,98 Subsequent to ischemia onset, there is an increase in extracellular ATP as a result of neuronal and glial depolarization and damaged plasma membrane of injured brain cells.93 High extracellular ATP levels can activate resident microglia and stimulate inflammatory cytokine production.93 Microglia and macrophages can assume a neurodegenerative M1 phenotype and increase inflammation by releasing proinflammatory cytokines. In the initial few minutes to hours after stroke onset, there is an increased production of proinflammatory cytokines (interleukins [eg, IL-6 and IL-1β), tumor necrosis factor-α, etc), integrins, adhesion molecules (VCAM-1 [vascular cell adhesion protein], ICAM-1 [intercellular adhesion molecule], P-selectin, etc), chemokines, and their receptor molecules.93,99,100 These proinflammatory molecules can further damage the BBB, initiate the recruitment of peripheral immune cells into the cerebral microcirculation and ischemic brain, and pass through the BBB into the circulation, inducing systemic inflammation.100 On the contrary, the microglia and macrophages can also assume an anti-inflammatory M2 phenotype and clear cellular debris from the injured brain.93
Systemic inflammatory responses are mainly driven by many cytokines, chemokines, stress hormones, and parasympathetic and sympathetic regulation.101 In acute ischemic stroke, lymphocyte influx begins ≈48 hours after stroke, and the invading T lymphocytes promote detrimental inflammatory cascades and induce delayed brain damage.97,99 Although an optimal-level leukocyte infiltration is beneficial to the ischemic brain, in excess, it leads to increased intracranial pressure resulting in poor outcome or mortality.93,99,100 The spleen plays a central role in mediating the peripheral immune response to ischemic stroke by increasing circulating lymphocytes and proinflammatory cytokines and chemokines such as tumor necrosis factor-α, MCP-1 (monocyte chemoattractant protein-1), interferon-γ, IL-6, and IL-2, exacerbating inflammation in the acute phase after stroke.102 Increased tumor necrosis factor-α level induces degradation of TnI, which has been associated with impaired cardiac contractility.103 Splenectomy 8 weeks after chronic heart failure has been reported to significantly improve LV systolic function and reduce cardiomyocyte hypertrophy, suggesting a role of spleen in mediating cardiac function.104 At a later time point, splenic atrophy and decrease in T-cell proliferation and inflammatory cytokines secretion lead to a state of immunosuppression.105
Damage-associated molecular patterns including various heat-shock proteins are released by dying brain cells after ischemic stroke.106 Injured astrocytes, neurons, and oligodendrocytes release brain-derived antigens such as glial fibrillary acidic protein, S100, and MBP (myelin basic protein).107 Damage-associated molecular patterns and brain-derived antigens can also pass the ruptured BBB and enter the systemic circulation. Damage-associated molecular patterns can activate TLRs (Toll-like receptors) and lead to cytokine and chemokine production.108 In experimental stroke, lymphocyte response to the brain-derived antigens, such as MBP, leads to antigen-specific secretion of transforming growth factor-β, consistent with a regulatory T cells’ response.109 However, in the presence of systemic inflammation, the antigen-specific autoimmune response is characterized by T-helper cells 1 and TLR activation and secretion of proinflammatory interferon-γ), IL-2, etc.110,111 TLR-4 is expressed on the cell surface of cardiomyocytes. TLR-4 activation and chronic inflammation with increased release of proinflammatory cytokines such as tumor necrosis factor-α IL-6, IL-1β have been implicated in developing heart failure and poor prognosis of heart disease.112 Transforming growth factor-β induces IL-6 increase while decreasing MCP-1 and VCAM-1 in vascular pericytes and is also instrumental in the transition from fibroblasts to myofibroblasts that may play a role in cardiac fibrosis observed in NSC.113,114
Systemic inflammatory responses are activated after spontaneous intracerebral hemorrhage, ischemic stroke, and SAH.115 Systemic inflammatory responses carry an increased risk of subsequent intracranial complications such as vasospasm and normal pressure hydrocephalus, as well as systemic secondary complications.116 The immune system is involved in this process in the early phases after stroke and correlates with ischemic stroke and SAH progression.116 Systemic inflammatory response syndrome was found in ≈50% of SAH patients on admission, and 85% of patients developed systemic inflammatory response syndrome within the first 4 days after admission.117 Systemic inflammatory response syndrome is indicated by abnormal leukocyte counts, high respiratory and heart rate (>90 bpm), and abnormal body temp and can increase the risk of developing vasospasm and secondary complications.117 In ischemic stroke patients, the number of CD74+ cells (the MHC [major histocompatibility complex] class II invariant chain present on all class II expressing cells, including monocytes, macrophages, and dendritic cells) were significantly increased in peripheral blood mononuclear cells compared with healthy controls.118 The increased CD74-positive cells were mainly on CD4+ T cells, monocytes, and dendritic cells.118 In heart diseases such as myocardial infarction, elevation of circulating blood monocytes is associated with poor cardiac function and predicts poor recovery of LV systolic function at 3 months post-injury.119
There is also some evidence of cross-talk between inflammatory responses and sympathetic activation in ischemic and hemorrhagic stroke.120,121 The proinflammatory cytokines released by damaged neuronal and glial cells stimulate the posterior hypothalamus to increase sympathetic output and circulating catecholamine level.122124 van der Bilt et al121 found that catecholaminergic stress after brain hemorrhage coincided with influx of inflammatory cells into the heart and induced cardiac damage after SAH. Neutrophils were found embedded in the inflamed myocardium along with thrombi.121 The coupling of catecholamine release with parasympathetic dysfunction regulates inflammation that induces myocardial dysfunction, thrombi formation, and cardiomyocyte cell death.121 Therefore, inflammation may play an important role and as a link between brain injury and cardiac damage after ischemic stroke and SAH. The role of immune response and inflammation after stroke in mediating cardiac dysfunction is summarized in Figure 3.
Gut microbiome dysbiosis after stroke can lead to intestinal paralysis and bacterial and endotoxin translocation to blood leading to systemic inflammation,125127 as discussed in detail in the following section.

Gut Microbiome Dysbiosis After Stroke

Our knowledge and understanding of the human microbiome and its involvement in various diseases are increasing. In particular, there is an emerging body of evidence based from experimental studies that suggests an interaction between the gut microbiome and heart as well as the gut microbiota and the CNS, referred to as gut–heart axis and brain–gut axis, respectively. The gut–blood barrier mainly regulates the absorption of nutrients and water while preventing toxins and pathogenic microorganisms from entering the blood stream. In gastrointestinal, metabolic, cardiovascular, and cerebrovascular diseases, disruption of the gut–blood barrier integrity resulting in increased intestinal permeability enables microbiota-derived molecules to enter the blood stream.125

Brain–Gut Axis

The brain–gut axis can affect normal brain functioning and influence the pathological cascade of events in neurological diseases including stroke and TBI.128,129 The brain–gut axis comprising neural and humoral pathways with signaling molecules such as cytokines, hormones, and neuropeptides can regulate immune response and lymphocyte population.129,130 Alterations to the normal gut microbiota or gut dysbiosis caused by acute brain lesions can affect neuroinflammatory and immune responses in the brain and worsen neurological function.131 In patients with large-artery atherosclerotic ischemic stroke or transient ischemic attack, significant gut dysbiosis is indicated by increased opportunistic bacteria and decreased beneficial bacteria.132 It has also been suggested that commensal gut microbiota exert protection against ischemic damage, and their depletion or dysbiosis increases post-stroke mortality in mice and influences stroke outcome.133 In addition, the extent of increase of proteobacteria in the gut of stroke patients is proportional to stroke severity.132 Dietary choline and l-carnitine are transformed by gut microbiota to trimethylamine, and its oxidation yields the proatherogenic metabolite that is largely considered a disease marker.134
In ischemic stroke, gut dysbiosis via stress-mediated intestinal paralysis can alter T-cell homeostasis, promote migration of T cells from the intestine to the ischemic brain, and increase proinflammatory responses resulting in poor stroke outcome.130 In mice subjected to transient focal cerebral ischemia, trafficking of intestinal T cells from the gut to the meninges of the brain increases neuroinflammation and the secretion of proinflammatory cytokine IL-17, which can stimulate the production of several other cytokines and chemokines and facilitate the infiltration of cytotoxic immune cells and neutrophils into the injured brain.130 In mice subjected to transient focal cerebral ischemia, stroke increased gut permeability proportional to stroke severity; promoted bacterial translocation from the gut to mesenteric lymph nodes and peripheral organs such as spleen, liver, and lungs; and triggered adaptive and innate immune responses.135 In ischemic stroke patients, gut dysbiosis and increased bacterial counts of Lactobacillus ruminis subgroup in the fecal gut microbiota was associated with elevated systemic inflammation and altered metabolism.136 Bacterial metabolites have also been implicated to mediate communication between the commensal gut microbiota and the immune system, tipping the balance in favor of proinflammatory mechanism.137

Gut–Heart Axis

Increased gut permeability can promote inflammatory responses, whereas systemic inflammation can increase gut permeability.125 Bacterial and endotoxin translocation to the blood stream, increased proinflammatory cytokines, and systemic inflammation can induce or exacerbate cardiac dysfunction.126,127 In an independent large clinical cohort, 3 gut microbiota–dependent metabolites including choline, trimethylamine-N-oxide, and betaine were found to be predictors of cardiovascular disease.134 Gut microbes can directly contribute to platelet hyper-reactivity and increase the risk of thrombosis via trimethylamine-N-oxide generation.138 Altered trimethylamine-N-oxide levels has been associated with impaired cardiac function, heart attack, and heart failure.126,138 Elevated trimethylamine-N-oxide levels in the plasma of patients presenting with chest pain was found to be a predictor of short-term and long-term risk of myocardial infarction, stroke, need for revascularization, or even death.139 Figure 4 summarizes the role of gut dysbiosis in mediating cardiac damage after stroke. Future studies are warranted to investigate the role of the gut microbiome in cardiac damage after stroke and investigate whether stroke-induced gut dysbiosis promotes a proinflammatory cascade, which then leads to dysfunction of other organs including the heart.
Figure 4. The brain–gut axis and gut–heart axis may mediate cardiac damage after stroke. Stroke increases intestinal barrier permeability, enabling bacterial and endotoxin translocation to bloodstream inducing inflammatory responses and proinflammatory cytokine production. Cytokine production can trigger inflammation, fibrosis, and microvascular and myocardial dysfunction. Trimethylamine-N-oxide (TMAO) is the hepatic oxidation product of the microbial metabolite trimethylamine (TMA). TMAO promotes the development of hyper-responsive platelet phenotype and enhances elevating the risk for heart failure and heart attack. CRP indicates C-reactive protein; IL, interleukin; and TNF, tumor necrosis factor.

Future Perspectives

Although there exist knowledge gaps in our understanding of the mechanisms of brain–heart interaction in the setting of stroke, emerging evidence suggests that microvesicles and their cargo miR may function as mediators of interorgan and intercellular communication. Important unanswered questions relating to the role of microvesicles/miR in brain–heart interaction include How do changes in miRs in the brain affect cardiac function and vice versa? Do changes in miR expression correspond to stages of disease and recovery? Are microvesicles containing miRs transported by the circulation from the brain to heart (as well as other organs), thereby inducing damage to target tissue? Because miRs can affect immune response, it is also possible that injured brain cells and resident and infiltrating inflammatory cells release a variety of microvesicles carrying miRs that on reaching cardiac tissue have adverse effects on cardiac function? The following section explores this perspective, as summarized in Figure 5. Further studies are warranted to elucidate the role of these intercellular communication mediators in facilitating interaction between the brain and distant organs such as the heart in neurological diseases.
Figure 5. The role of microvesicles (MVs) released by damaged astrocytes, neurons, and microglia after stroke in mediating cardiac damage warrants future investigation. Stroke-induced elevation of circulating platelets and MVs are associated with endothelial dysfunction and coagulation leading to thrombosis. MVs induce endothelial dysfunction by decreasing NO synthesis as the result of endothelial NO synthase (eNOS) function inhibition and an increase in caveolin-1. The outer leaflet of the MVs membrane contains 2 procoagulants: phosphatidylserine and tissue factor. MVs can bind to coagulation factors and promote their activation. In addition, MVs harbor tissue factor, which can initiate extrinsic coagulation pathways and promote the assembly of clotting enzymes leading to thrombin generation. At sites of vascular injury, P-selectin exposure by activated endothelial cells or platelets leads to the rapid recruitment of MVs bearing the P-selectin glycoprotein ligand-1 (PSGL-1). The interactions between PSGL-1 and platelet P-selectin are necessary to concentrate tissue factor activity at the thrombus.

Microvesicles

Microvesicles are lipid bilayer-covered cell fragments that range in diameter from 100 nm to 1 µm and are released into the extracellular environment by several cell types typically during cell activation or cell death via necrosis or apoptosis. Microvesicles are produced by the outward budding and fission of plasma membrane. In addition to intercellular communication via cell contact, microvesicles constitute an important mode of communication by serving as vehicles for intercellular transfer of membrane and cytosolic proteins, lipids, and RNA either locally or over long distances.140 Because microvesicles retain the signature membrane protein composition of the parent cell, they can be classified by flow cytometry into different cellular microvesicles, such as endothelial microvesicles, leukocyte microvesicles, and platelet microvesicles or brain-derived microvesicles. Microvesicle shedding can also be triggered by pathological activation of inflammatory processes and activation of coagulation or complement systems or even by shear stress in the circulation. Microvesicles have a bilayered phospholipid structure exposing coagulant-active phosphatidylserine and expressing various membrane receptors, and they serve as cell-to-cell shuttles for bioactive molecules such as lipids, growth factors, miR, and mitochondria.140 Microvesicles have been implicated in endothelial dysfunction, inflammation, and thrombosis.141 In addition, microvesicles may also be targeted as a therapeutic delivery tool by either inhibiting their release from diseased cells or manipulating their cargo to enable shuttling of secretory molecules such as cytokines, chemokines, and growth factors.142

Circulating Microvesicles

Many circulating microvesicles in the blood derive from cells that are in contact with the bloodstream such as platelets and endothelial cells. Circulating microvesicle level is elevated in patients with ischemic stroke, intracerebral hemorrhage stroke, and SAH and is associated with poor clinical outcome.143,144 A major source of microvesicles in stroke patients is vascular endothelial cells.145 Blood-derived microvesicles stimulate release of endothelial cell cytokine IL-6.145 Correlation between IL-6 expression and incidence of vasospasm has been demonstrated previously.146 Therefore, in peripheral blood vessels, including cardiac vasculature, IL-6 can induce vascular spasm and lead to coronary syndrome. In addition, endothelial cells secrete microvesicles that can inhibit endothelial NO synthase.147 Anti–endothelial NO synthase activity leads to a decline in NO production, causing endothelial dysfunction and impaired vascular relaxation.148,149 Endothelial microvesicle was significantly increased in SAH patients, and the increased endothelial cell microvesicle is related with symptomatic cerebral vasospasm143 and predicts infarction post-SAH.150 Microvesicles may play an important role for secondary complications in patients with ischemic and hemorrhagic stroke.151
Previous studies have demonstrated that platelet microvesicles have 50- to 100-fold higher specific procoagulant activity than activated platelets152 and are hence associated with thrombosis. Platelets play an important role in maintaining hemostasis such as thrombosis and immune response. Platelets are activated by inflammation, infection, or injury, and after their activation, microvesicles are released from platelets. In intracerebral hemorrhage, cerebrospinal fluid and plasma procoagulant MV levels are significantly increased and are related with stroke pathogenesis.144 Platelet microvesicles may also serve as a link between vascular coagulation and inflammation in cardiovascular disease.153 Altered platelet activity because of microvesicles derived from platelets and injured brain can lead to thromboembolic events and associated cardiac complications after brain injury such as stroke or TBI.153 Circulating microvesicles derived from brain, endothelial cells, and blood cells may promote procoagulant activity and thrombin generation.153 However, microvesicles can contribute to the onset and progression of some neurodegenerative and neuroinflammatory diseases, as well as to the development and regeneration of the nervous system. Activated platelets shed microvesicles, which contain a variety of growth factors augmenting endogenous neural progenitor and stem cells angiogenesis and neurogenesis, which may be used for therapy for ischemic stroke.154

Neural Source of Microvesicles

Neurons, astrocytes, microglia, and neural stem cells release microvesicles under normal and pathological conditions. Neural microvesicles can contribute to the onset and progression of some neurodegenerative and neuroinflammatory diseases and to the development and regeneration of the nervous system after stroke.155 Microglial microvesicles and astrocyte microvesicles store and release the inflammatory cytokine IL-1β.156,157 In addition, microvesicles contribute to IL-1β release from glial cells.157 In the acute phase of injury after TBI in mice, circulating platelet numbers did not change; however, platelet contribution to clot formation and the number of total circulating microvesicles decreased significantly.158 When these TBI mice were treated with microvesicles derived from the brain tissue of sham mice, platelet contribution to clot formation normalized.158 This indicates that brain tissue–secreted microvesicles generated after TBI are likely to mediate post-traumatic hypercoagulable state after TBI.158 Tian et al found that microvesicles were produced from injured hippocampal cells, transmigrated through the disrupted endothelial barrier in a platelet-dependent manner, and activated platelets and thereby induce coagulopathy in TBI model.158 Zhao et al159 found that brain-derived mitochondrial microparticles significantly increased in the circulation after TBI. The mitochondrial microparticles synergized with platelets to facilitate vascular leakage by disrupting the endothelial barrier. The disrupted endothelial barrier allowed the release of mitochondrial microparticles into the systemic circulation to promote coagulation and enhanced fibrinolysis, vascular fibrin deposition, and thrombosis.159 The underlying mechanisms and role played by microvesicles in mediating cardiac complications after cerebral injury are emerging and warrant further study. Increased coagulation and thrombosis may induce heart damage after brain injury.

MicroRNA

MiRs are short sequences of noncoding RNA (ca. 22 nucleotides), which regulate gene expression both transcriptionally and post-transcriptionally.160 MiRs can regulate several genes, pathways, and biological networks, either acting alone or in concert with other miRs. MiRs regulate many biological processes regulating tissue repair, including angiogenesis, inflammation, and hypoxia–response.160 About 50 circulating miRs are believed to be associated with cardiovascular diseases including miR-1, miR-16, miR-27b, miR-30d, miR-126, miR-133, miR-143, miR-145, miR-208, and the let-7 family.161 Several miRs with key cardiac and vascular function have been reported to be affected after stroke including the following: miR-23, miR-24, miR-29, miR-30, miR-103, and miR-222 were upregulated, whereas miR-126 was downregulated.162 Microvesicles also contain abundant miRs.
Endothelial cell–specific miR-126 has a key role in maintaining vascular integrity and regulating angiogenesis.163 Circulating miR-126 level is significantly decreased in patients with ischemic stroke until at least 24 weeks.164 Endothelial cell dysfunction is a principal step in atherosclerosis. MiR-126 deficiency has been highly associated with heart failure, atrial fibrillation, and coronary artery disease and may be associated with stroke-induced severe cardiac complications.163,165 Coronary artery disease or ischemic heart disease includes stable and unstable angina, myocardial infarction, and sudden cardiac death. In myocardial infarction, circulating miR-1 increases and miR-126 decreases in proportion with cTnI plasma concentration levels.166 Vascular miR-126 has been demonstrated to be consumed or taken up by the heart from the bloodstream according to a transcoronary gradient.167 It has been demonstrated that ischemic stroke decreases serum and heart miR-126 expression, as well as induces post-stroke cardiac dysfunction.168 Conditional specific endothelial cell miR-126 knockout mice exhibited enhanced cardiac dysfunction and increased cardiomyocyte hypertrophy, fibrosis, and inflammatory factor expression after ischemic stroke compared with miR-126 knockout control stroke mice.168 The study demonstrated that decreased miR-126 may play a role in brain–heart interaction after ischemic stroke.168
In addition, circulating miR-145 also significantly increases within 24 hours of cerebral ischemia, and circulating miR-145 level is positively correlated with elevated serum inflammatory factor IL-6.169 MiR-145 modulates endothelial cell function in angiogenesis and vessel stabilization.170 Circulating miR-145 changes has also been reported in patients with coronary artery disease and acute myocardial infarction.171 Therefore, other miRs may also regulate brain–heart interaction.

Conclusions

In summary, brain–heart syndrome is commonly encountered in the clinic and affects the prognosis, morbidity, and mortality of patients. The secondary brain damage caused by cardiac dysfunction and impaired systemic and brain homeostasis is a serious problem that requires attention. Although there is a necessity for specific interventions for the prevention and treatment of post-stroke cardiac complications, there is a dearth of data to guide the management of these complications. Investigation of clinical indicators to diagnose the adverse brain–heart disease at early stage will enable improved clinical management and reduce mortality. Research of novel therapies for the protection of heart after stroke warrants investigation with the potential to be applied to a large proportion of stroke patients over a wide time window.

Footnote

Nonstandard Abbreviations and Acronyms

BBB
blood–brain barrier
CNS
central nervous system
cTnI
cardiac troponin I
cTnT
Cardiac Troponin T
HF
high frequency
HPA
hypothalamic pituitary adrenal
HRV
heart rate variability
hsTnT
high-sensitivity troponin T
IL
interleukin
LF
low frequency
MBP
myelin basic protein
MCP-1
monocyte chemoattractant protein-1
MiR
MicroRNA
NSC
Neurogenic stress cardiomyopathy
NT-proBNP
N-terminal fragment of B-type natriuretic peptide
SAH
Subarachnoid hemorrhage
TBI
traumatic brain injury
TLRs
Toll-like receptors

References

1.
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. doi: 10.1212/01.wnl.0000206077.13705.6d.
2.
Oppenheimer SM. Neurogenic cardiac effects of cerebrovascular disease. Curr Opin Neurol. 1994;7:20–24.
3.
Tokgözoglu SL, Batur MK, Topçuoglu MA, Saribas O, Kes S, Oto A. Effects of stroke localization on cardiac autonomic balance and sudden death. Stroke. 1999;30:1307–1311.
4.
Byer E, Ashman R, Toth LA. Electrocardiograms with large, upright T waves and long Q-T intervals. Am Heart J. 1947;33:796–806.
5.
Samuels MA. The brain-heart connection. Circulation. 2007;116:77–84. doi: 10.1161/CIRCULATIONAHA.106.678995.
6.
Tranmer BI, Keller TS, Kindt GW, Archer D. Loss of cerebral regulation during cardiac output variations in focal cerebral ischemia. J Neurosurg. 1992;77:253–259.
7.
Cheshire WP, Saper CB. The insular cortex and cardiac response to stroke. Neurology. 2006;66:1296–1297. doi: 10.1212/01.wnl.0000219563.87204.7d.
8.
van der Bilt IA, Hasan D, Vandertop WP, Wilde AA, Algra A, Visser FC, Rinkel GJ. Impact of cardiac complications on outcome after aneurysmal subarachnoid hemorrhage: a meta-analysis. Neurology. 2009;72:635–642. doi: 10.1212/01.wnl.0000342471.07290.07.
9.
Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham Study. Stroke. 1991;22:983–988.
10.
Verdecchia P, Porcellati C, Reboldi G, Gattobigio R, Borgioni C, Pearson TA, Ambrosio G. Left ventricular hypertrophy as an independent predictor of acute cerebrovascular events in essential hypertension. Circulation. 2001;104:2039–2044.
11.
Selvetella G, Notte A, Maffei A, Calistri V, Scamardella V, Frati G, Trimarco B, Colonnese C, Lembo G. Left ventricular hypertrophy is associated with asymptomatic cerebral damage in hypertensive patients. Stroke. 2003;34:1766–1770. doi: 10.1161/01.STR.0000078310.98444.1D.
12.
Petersen P. Thromboembolic complications in atrial fibrillation. Stroke. 1990;21:4–13.
13.
Khechinashvili G, Asplund K. Electrocardiographic changes in patients with acute stroke: a systematic review. Cerebrovasc Dis. 2002;14:67–76.
14.
Mosterd A, Hoes AW. Clinical epidemiology of heart failure. Heart. 2007;93:1137–1146. doi: 10.1136/hrt.2003.025270.
15.
Yoshimura S, Toyoda K, Ohara T, Nagasawa H, Ohtani N, Kuwashiro T, Naritomi H, Minematsu K. Takotsubo cardiomyopathy in acute ischemic stroke. Ann Neurol. 2008;64:547–554. doi: 10.1002/ana.21459.
16.
van der Bilt IA, Hasan D, van den Brink RB, et al.; SEASAH (Serial Echocardiography After Subarachnoid Hemorrhage) Investigators. Time course and risk factors for myocardial dysfunction after aneurysmal subarachnoid hemorrhage. Neurosurgery. 2015;76:700–705; discussion 705. doi: 10.1227/NEU.0000000000000699.
17.
Ghadri JR, Cammann VL, Napp LC, et al.; International Takotsubo (InterTAK) Registry. Differences in the clinical profile and outcomes of typical and atypical takotsubo syndrome: data from the International Takotsubo Registry. JAMA Cardiol. 2016;1:335–340. doi: 10.1001/jamacardio.2016.0225.
18.
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. doi: 10.1159/000135711.
19.
Estanol BV, Marin OS. Cardiac arrhythmias and sudden death in subarachnoid hemorrhage. Stroke. 1975;6:382–386.
20.
Junttila E, Vaara M, Koskenkari J, Ohtonen P, Karttunen A, Raatikainen P, Ala-Kokko T. Repolarization abnormalities in patients with subarachnoid and intracerebral hemorrhage: predisposing factors and association with outcome. Anesth Analg. 2013;116:190–197. doi: 10.1213/ANE.0b013e318270034a.
21.
Jangra K, Grover VK, Bhagat H, Bhardwaj A, Tewari MK, Kumar B, Panda NB, Sahu S. Evaluation of the effect of aneurysmal clipping on electrocardiography and echocardiographic changes in patients with subarachnoid hemorrhage: a prospective observational study. J Neurosurg Anesthesiol. 2017;29:335–340. doi: 10.1097/ANA.0000000000000318.
22.
Lee M, Oh JH, Lee KB, Kang GH, Park YH, Jang WJ, Chun WJ, Lee SH, Lee IC. Clinical and echocardiographic characteristics of acute cardiac dysfunction associated with acute brain hemorrhage- difference from takotsubo cardiomyopathy. Circ J. 2016;80:2026–2032. doi: 10.1253/circj.CJ-16-0395.
23.
Hays A, Diringer MN. Elevated troponin levels are associated with higher mortality following intracerebral hemorrhage. Neurology. 2006;66:1330–1334. doi: 10.1212/01.wnl.0000210523.22944.9b.
24.
Park HK, 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. doi: 10.5853/jos.2015.01697.
25.
Milionis H, Faouzi M, Cordier M, D’Ambrogio-Remillard S, Eskandari A, Michel P. Characteristics and early and long-term outcome in patients with acute ischemic stroke and low ejection fraction. Int J Cardiol. 2013;168:1082–1087. doi: 10.1016/j.ijcard.2012.11.036.
26.
Kolin A, Norris JW. Myocardial damage from acute cerebral lesions. Stroke. 1984;15:990–993.
27.
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. doi: 10.1161/STROKEAHA.106.471813.
28.
Rauh R, Fischereder M, Spengel FA. Transesophageal echocardiography in patients with focal cerebral ischemia of unknown cause. Stroke. 1996;27:691–694.
29.
Lavy S, Yaar I, Melamed E, Stern S. The effect of acute stroke on cardiac functions as observed in an intensive stroke care unit. Stroke. 1974;5:775–780.
30.
McDermott MM, Lefevre F, Arron M, Martin GJ, Biller J. ST segment depression detected by continuous electrocardiography in patients with acute ischemic stroke or transient ischemic attack. Stroke. 1994;25:1820–1824.
31.
Ay H, Arsava EM, Saribaş O. Creatine kinase-MB elevation after stroke is not cardiac in origin: comparison with troponin T levels. Stroke. 2002;33:286–289.
32.
Hall TS, Hallén J, Krucoff MW, Roe MT, Brennan DM, Agewall S, Atar D, Lincoff AM. Cardiac troponin I for prediction of clinical outcomes and cardiac function through 3-month follow-up after primary percutaneous coronary intervention for ST-segment elevation myocardial infarction. Am Heart J. 2015;169:257–265.e1. doi: 10.1016/j.ahj.2014.10.015.
33.
Apple FS, Steffen LM, Pearce LA, Murakami MM, Luepker RV. Increased cardiac troponin I as measured by a high-sensitivity assay is associated with high odds of cardiovascular death: the Minnesota Heart Survey. Clin Chem. 2012;58:930–935. doi: 10.1373/clinchem.2011.179176.
34.
Kubo T, Kitaoka H, Okawa M, Yamanaka S, Hirota T, Baba Y, Hayato K, Yamasaki N, Matsumura Y, Yasuda N, Sugiura T, Doi YL. Combined measurements of cardiac troponin I and brain natriuretic peptide are useful for predicting adverse outcomes in hypertrophic cardiomyopathy. Circ J. 2011;75:919–926.
35.
Bugnicourt JM, Rogez V, Guillaumont MP, Rogez JC, Canaple S, Godefroy O. Troponin levels help predict new-onset atrial fibrillation in ischaemic stroke patients: a retrospective study. Eur Neurol. 2010;63:24–28. doi: 10.1159/000258679.
36.
Oras J, Grivans C, Bartley A, Rydenhag B, Ricksten SE, Seeman-Lodding H. Elevated high-sensitive troponin T on admission is an indicator of poor long-term outcome in patients with subarachnoid haemorrhage: a prospective observational study. Crit Care. 2016;20:11. doi: 10.1186/s13054-015-1181-5.
37.
Bruder N, Rabinstein A; Participants in the International Multi-Disciplinary Consensus Conference on the Critical Care Management of Subarachnoid Hemorrhage. Cardiovascular and pulmonary complications of aneurysmal subarachnoid hemorrhage. Neurocrit Care. 2011;15:257–269. doi: 10.1007/s12028-011-9598-4.
38.
Garrett MC, Komotar RJ, Starke RM, Doshi D, Otten ML, Connolly ES. Elevated troponin levels are predictive of mortality in surgical intracerebral hemorrhage patients. Neurocrit Care. 2010;12:199–203. doi: 10.1007/s12028-009-9245-5.
39.
Di Angelantonio E, Fiorelli M, Toni D, Sacchetti ML, Lorenzano S, Falcou A, Ciarla MV, Suppa M, Bonanni L, Bertazzoni G, Aguglia F, Argentino C. Prognostic significance of admission levels of troponin I in patients with acute ischaemic stroke. J Neurol Neurosurg Psychiatry. 2005;76:76–81. doi: 10.1136/jnnp.2004.041491.
40.
Cushman M, Judd SE, Howard VJ, Kissela B, Gutiérrez OM, Jenny NS, Ahmed A, Thacker EL, Zakai NA. N-terminal pro-B-type natriuretic peptide and stroke risk: the reasons for geographic and racial differences in stroke cohort. Stroke. 2014;45:1646–1650. doi: 10.1161/STROKEAHA.114.004712.
41.
Krause T, Werner K, Fiebach JB, Villringer K, Piper SK, Haeusler KG, Endres M, Scheitz JF, Nolte CH. Stroke in right dorsal anterior insular cortex Is related to myocardial injury. Ann Neurol. 2017;81:502–511. doi: 10.1002/ana.24906.
42.
Nigro N, Wildi K, Mueller C, Schuetz P, Mueller B, Fluri F, Christ-Crain M, Katan M. BNP but Not s-cTnln is associated with cardioembolic aetiology and predicts short and long term prognosis after cerebrovascular events. PLoS One. 2014;9:e102704. doi: 10.1371/journal.pone.0102704.
43.
Yip HK, Sun CK, Chang LT, Chen MC, Liou CW. Time course and prognostic value of plasma levels of N-terminal pro-brain natriuretic peptide in patients after ischemic stroke. Circ J. 2006;70:447–452.
44.
Etgen T, Baum H, Sander K, Sander D. Cardiac troponins and N-terminal pro-brain natriuretic peptide in acute ischemic stroke do not relate to clinical prognosis. Stroke. 2005;36:270–275. doi: 10.1161/01.STR.0000151364.19066.a1.
45.
Tung P, Kopelnik A, Banki N, Ong K, Ko N, Lawton MT, Gress D, Drew B, Foster E, Parmley W, Zaroff J. Predictors of neurocardiogenic injury after subarachnoid hemorrhage. Stroke. 2004;35:548–551. doi: 10.1161/01.STR.0000114874.96688.54.
46.
Jauch EC, Saver JL, Adams HP, Bruno A, Connors JJ, Demaerschalk BM, Khatri P, McMullan PW, Qureshi AI, Rosenfield K, Scott PA, Summers DR, Wang DZ, Wintermark M, Yonas H; American Heart Association Stroke Council; Council on Cardiovascular Nursing; Council on Peripheral Vascular Disease; Council on Clinical Cardiology. Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2013;44:870–947. doi: 10.1161/STR.0b013e318284056a.
47.
Billman GE. The LF/HF ratio does not accurately measure cardiac sympatho-vagal balance. Front Physiol. 2013;4:26. doi: 10.3389/fphys.2013.00026.
48.
Papaioannou V, Pneumatikos I, Maglaveras N. Association of heart rate variability and inflammatory response in patients with cardiovascular diseases: current strengths and limitations. Front Physiol. 2013;4:174. doi: 10.3389/fphys.2013.00174.
49.
Korpelainen JT, Sotaniemi KA, Huikuri HV, Myllyä VV. Abnormal heart rate variability as a manifestation of autonomic dysfunction in hemispheric brain infarction. Stroke. 1996;27:2059–2063.
50.
Korpelainen JT, Sotaniemi KA, Mäkikallio A, Huikuri HV, Myllylä VV. Dynamic behavior of heart rate in ischemic stroke. Stroke. 1999;30:1008–1013.
51.
Chen WL, Huang CH, Chen JH, Tai HC, Chang SH, Wang YC. ECG abnormalities predict neurogenic pulmonary edema in patients with subarachnoid hemorrhage. Am J Emerg Med. 2016;34:79–82. doi: 10.1016/j.ajem.2015.09.032.
52.
Colivicchi F, Bassi A, Santini M, Caltagirone C. Cardiac autonomic derangement and arrhythmias in right-sided stroke with insular involvement. Stroke. 2004;35:2094–2098. doi: 10.1161/01.STR.0000138452.81003.4c.
53.
Naver HK, Blomstrand C, Wallin BG. Reduced heart rate variability after right-sided stroke. Stroke. 1996;27:247–251.
54.
Loubinoux I, Kronenberg G, Endres M, Schumann-Bard P, Freret T, Filipkowski RK, Kaczmarek L, Popa-Wagner A. Post-stroke depression: mechanisms, translation and therapy. J Cell Mol Med. 2012;16:1961–1969. doi: 10.1111/j.1582-4934.2012.01555.x.
55.
Hachinski V. Post-stroke depression, not to be underestimated. Lancet. 1999;353:1728. doi: 10.1016/S0140-6736(99)00139-7.
56.
Rosmond R, Björntorp P. The hypothalamic-pituitary-adrenal axis activity as a predictor of cardiovascular disease, type 2 diabetes and stroke. J Intern Med. 2000;247:188–197.
57.
Whitnall MH. Regulation of the hypothalamic corticotropin-releasing hormone neurosecretory system. Prog Neurobiol. 1993;40:573–629.
58.
Christensen H, Boysen G, Johannesen HH. Serum-cortisol reflects severity and mortality in acute stroke. J Neurol Sci. 2004;217:175–180.
59.
Barugh AJ, Gray P, Shenkin SD, MacLullich AM, Mead GE. Cortisol levels and the severity and outcomes of acute stroke: a systematic review. J Neurol. 2014;261:533–545. doi: 10.1007/s00415-013-7231-5.
60.
Melville KI, Blum B, Shister HE, Silver MD. Cardiac ischemic changes and arrhythmias induced by hypothalamic stimulation. Am J Cardiol. 1963;12:781–791.
61.
Jia S, Xia Q, Zhang B, Wang L. Involvement of the paraventricular nucleus in the occurrence of arrhythmias in middle cerebral artery occlusion rats. J Stroke Cerebrovasc Dis. 2015;24:844–851. doi: 10.1016/j.jstrokecerebrovasdis.2014.11.025.
62.
Infanger DW, Cao X, Butler SD, Burmeister MA, Zhou Y, Stupinski JA, Sharma RV, Davisson RL. Silencing nox4 in the paraventricular nucleus improves myocardial infarction-induced cardiac dysfunction by attenuating sympathoexcitation and periinfarct apoptosis. Circ Res. 2010;106:1763–1774. doi: 10.1161/CIRCRESAHA.109.213025.
63.
Fassbender K, Schmidt R, Mössner R, Daffertshofer M, Hennerici M. Pattern of activation of the hypothalamic-pituitary-adrenal axis in acute stroke. Relation to acute confusional state, extent of brain damage, and clinical outcome. Stroke. 1994;25:1105–1108.
64.
Samuels MA. Neurogenic heart disease: a unifying hypothesis. Am J Cardiol. 1987;60:15J–19J.
65.
Schömig A. Catecholamines in myocardial ischemia. Systemic and cardiac release. Circulation. 1990;82:II13–II22.
66.
Mertes PM, Carteaux JP, Jaboin Y, Pinelli G, el Abassi K, Dopff C, Atkinson J, Villemot JP, Burlet C, Boulange M. Estimation of myocardial interstitial norepinephrine release after brain death using cardiac microdialysis. Transplantation. 1994;57:371–377.
67.
Jacob WA, Van Bogaert A, De Groodt-Lasseel MH. Myocardial ultrastructure and haemodynamic reactions during experimental subarachnoid haemorrhage. J Mol Cell Cardiol. 1972;4:287–298.
68.
Costa VM, Carvalho F, Bastos ML, Carvalho RA, Carvalho M, Remião F. Contribution of catecholamine reactive intermediates and oxidative stress to the pathologic features of heart diseases. Curr Med Chem. 2011;18:2272–2314.
69.
Eitel I, von Knobelsdorff-Brenkenhoff F, Bernhardt P, Carbone I, Muellerleile K, Aldrovandi A, Francone M, Desch S, Gutberlet M, Strohm O, Schuler G, Schulz-Menger J, Thiele H, Friedrich MG. Clinical characteristics and cardiovascular magnetic resonance findings in stress (takotsubo) cardiomyopathy. JAMA. 2011;306:277–286. doi: 10.1001/jama.2011.992.
70.
Nef HM, Möllmann H, Hilpert P, Troidl C, Voss S, Rolf A, Behrens CB, Weber M, Hamm CW, Elsässer A. Activated cell survival cascade protects cardiomyocytes from cell death in Tako-Tsubo cardiomyopathy. Eur J Heart Fail. 2009;11:758–764. doi: 10.1093/eurjhf/hfp076.
71.
Hachinski VC, Smith KE, Silver MD, Gibson CJ, Ciriello J. Acute myocardial and plasma catecholamine changes in experimental stroke. Stroke. 1986;17:387–390.
72.
Weinberg SJ, Fuster JM. Electrocardiographic changes produced by localized hypothalamic stimulations. Ann Intern Med. 1960;53:332–341.
73.
Cruickshank JM, Neil-Dwyer G, Stott AW. Possible role of catecholamines, corticosteroids, and potassium in production of electrocardiographic abnormalities associated with subarachnoid haemorrhage. Br Heart J. 1974;36:697–706.
74.
Moss RL, Fitzsimons DP, Ralphe JC. Cardiac MyBP-C regulates the rate and force of contraction in mammalian myocardium. Circ Res. 2015;116:183–192. doi: 10.1161/CIRCRESAHA.116.300561.
75.
Saini HK, Tripathi ON, Zhang S, Elimban V, Dhalla NS. Involvement of Na+/Ca2+ exchanger in catecholamine-induced increase in intracellular calcium in cardiomyocytes. Am J Physiol Heart Circ Physiol. 2006;290:H373–H380. doi: 10.1152/ajpheart.00613.2005.
76.
Hunt D, Gore I. Myocardial lesions following experimental intracranial hemorrhage: prevention with propranolol. Am Heart J. 1972;83:232–236.
77.
Madias JE. What is the recurrence rate of takotsubo syndrome in patients treated with β-blockers and angiotensin converting enzyme inhibitors/angiotensin receptor blockers? Int J Cardiol. 2016;219:394–395. doi: 10.1016/j.ijcard.2016.06.119.
78.
Tahsili-Fahadan P, Geocadin RG. Heart- brain axis: effects of neurologic injury on cardiovascular function. Circ Res. 2017;120:559–572. doi: 10.1161/CIRCRESAHA.116.308446.
79.
Hall RE, Livingston RB, Bloor CM. Orbital cortical influences on cardiovascular dynamics and myocardial structure in conscious monkeys. J Neurosurg. 1977;46:648–653.
80.
Sörös P, Hachinski V. Cardiovascular and neurological causes of sudden death after ischaemic stroke. Lancet Neurol. 2012;11:179–188. doi: 10.1016/S1474-4422(11)70291-5.
81.
Sander D, Winbeck K, Klingelhöfer J, Etgen T, Conrad B. Prognostic relevance of pathological sympathetic activation after acute thromboembolic stroke. Neurology. 2001;57:833–838.
82.
Ozdemir O, Hachinski V. Brain lateralization and sudden death: its role in the neurogenic heart syndrome. J Neurol Sci. 2008;268:6–11. doi: 10.1016/j.jns.2007.11.009.
83.
Min J, Farooq MU, Greenberg E, Aloka F, Bhatt A, Kassab M, Morgan JP, Majid A. Cardiac dysfunction after left permanent cerebral focal ischemia: the brain and heart connection. Stroke. 2009;40:2560–2563. doi: 10.1161/STROKEAHA.108.536086.
84.
Christensen H, Boysen G, Christensen AF, Johannesen HH. Insular lesions, ECG abnormalities, and outcome in acute stroke. J Neurol Neurosurg Psychiatry. 2005;76:269–271. doi: 10.1136/jnnp.2004.037531.
85.
Laowattana S, Zeger SL, Lima JA, Goodman SN, Wittstein IS, Oppenheimer SM. Left insular stroke is associated with adverse cardiac outcome. Neurology. 2006;66:477–483; discussion 463. doi: 10.1212/01.wnl.0000202684.29640.60.
86.
Algra A, Gates PC, Fox AJ, Hachinski V, Barnett HJ; North American Symptomatic Carotid Endarterectomy Trial Group. Side of brain infarction and long-term risk of sudden death in patients with symptomatic carotid disease. Stroke. 2003;34:2871–2875. doi: 10.1161/01.STR.0000099964.34430.2D.
87.
Masuda T, Sato K, Yamamoto S, Matsuyama N, Shimohama T, Matsunaga A, Obuchi S, Shiba Y, Shimizu S, Izumi T. Sympathetic nervous activity and myocardial damage immediately after subarachnoid hemorrhage in a unique animal model. Stroke. 2002;33:1671–1676.
88.
Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010;37:13–25. doi: 10.1016/j.nbd.2009.07.030.
89.
Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis. 2008;32:200–219. doi: 10.1016/j.nbd.2008.08.005.
90.
Varatharaj A, Galea I. The blood-brain barrier in systemic inflammation. Brain Behav Immun. 2017;60:1–12. doi: 10.1016/j.bbi.2016.03.010.
91.
Merino JG, Latour LL, Tso A, Lee KY, Kang DW, Davis LA, Lazar RM, Horvath KA, Corso PJ, Warach S. Blood-brain barrier disruption after cardiac surgery. AJNR Am J Neuroradiol. 2013;34:518–523. doi: 10.3174/ajnr.A3251.
92.
Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010;87:779–789. doi: 10.1189/jlb.1109766.
93.
Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med. 2011;17:796–808. doi: 10.1038/nm.2399.
94.
Gauberti M, Montagne A, Quenault A, Vivien D. Molecular magnetic resonance imaging of brain-immune interactions. Front Cell Neurosci. 2014;8:389. doi: 10.3389/fncel.2014.00389.
95.
Doll DN, Barr TL, Simpkins JW. Cytokines: their role in stroke and potential use as biomarkers and therapeutic targets. Aging Dis. 2014;5:294–306. doi: 10.14336/AD.2014.0500294.
96.
Dickens AM, Tovar-y-Romo LB, Yoo S-W, Trout AL, Bae M, Kanmogne M, Megra B, Williams DW, Witwer KW, Gacias M, Tabatadze N, Cole RN, Casaccia P, Berman JW, Anthony DC, Haughey NJ. Astrocyte-shed extracellular vesicles regulate the peripheral leukocyte response to inflammatory brain lesions. Sci Signal. 2017;10:pii-eaai7696. doi: 10.1126/scisignal.aai7696.
97.
Emsley HC, Smith CJ, Gavin CM, Georgiou RF, Vail A, Barberan EM, Hallenbeck JM, del Zoppo GJ, Rothwell NJ, Tyrrell PJ, Hopkins SJ. An early and sustained peripheral inflammatory response in acute ischaemic stroke: relationships with infection and atherosclerosis. J Neuroimmunol. 2003;139:93–101.
98.
Gelderblom M, Leypoldt F, Steinbach K, Behrens D, Choe CU, Siler DA, Arumugam TV, Orthey E, Gerloff C, Tolosa E, Magnus T. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke. 2009;40:1849–1857. doi: 10.1161/STROKEAHA.108.534503.
99.
Liesz A, Zhou W, Mracskó É, Karcher S, Bauer H, Schwarting S, Sun L, Bruder D, Stegemann S, Cerwenka A, Sommer C, Dalpke AH, Veltkamp R. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain. 2011;134:704–720. doi: 10.1093/brain/awr008.
100.
Yilmaz G, Granger DN. Leukocyte recruitment and ischemic brain injury. Neuromolecular Med. 2010;12:193–204. doi: 10.1007/s12017-009-8074-1.
101.
An C, Shi Y, Li P, Hu X, Gan Y, Stetler RA, Leak RK, Gao Y, Sun BL, Zheng P, Chen J. Molecular dialogs between the ischemic brain and the peripheral immune system: dualistic roles in injury and repair. Prog Neurobiol. 2014;115:6–24. doi: 10.1016/j.pneurobio.2013.12.002.
102.
Offner H, Subramanian S, Parker SM, Afentoulis ME, Vandenbark AA, Hurn PD. Experimental stroke induces massive, rapid activation of the peripheral immune system. J Cereb Blood Flow Metab. 2006;26:654–665. doi: 10.1038/sj.jcbfm.9600217.
103.
Adams V, Linke A, Wisloff U, Döring C, Erbs S, Kränkel N, Witt CC, Labeit S, Müller-Werdan U, Schuler G, Hambrecht R. Myocardial expression of Murf-1 and MAFbx after induction of chronic heart failure: effect on myocardial contractility. Cardiovasc Res. 2007;73:120–129. doi: 10.1016/j.cardiores.2006.10.026.
104.
Ismahil MA, Hamid T, Bansal SS, Patel B, Kingery JR, Prabhu SD. Remodeling of the mononuclear phagocyte network underlies chronic inflammation and disease progression in heart failure: critical importance of the cardiosplenic axis. Circ Res. 2014;114:266–282. doi: 10.1161/CIRCRESAHA.113.301720.
105.
Offner H, Subramanian S, Parker SM, Wang C, Afentoulis ME, Lewis A, Vandenbark AA, Hurn PD. Splenic atrophy in experimental stroke is accompanied by increased regulatory T cells and circulating macrophages. J Immunol. 2006;176:6523–6531.
106.
Shichita T, Hasegawa E, Kimura A, et al. Peroxiredoxin family proteins are key initiators of post-ischemic inflammation in the brain. Nat Med. 2012;18:911–917. doi: 10.1038/nm.2749.
107.
Becker K. Autoimmune responses to brain following stroke. Transl Stroke Res. 2012;3:310–317. doi: 10.1007/s12975-012-0154-0.
108.
Marsh BJ, Williams-Karnesky RL, Stenzel-Poore MP. Toll-like receptor signaling in endogenous neuroprotection and stroke. Neuroscience. 2009;158:1007–1020. doi: 10.1016/j.neuroscience.2008.07.067.
109.
Becker KJ, Kindrick DL, Lester MP, Shea C, Ye ZC. Sensitization to brain antigens after stroke is augmented by lipopolysaccharide. J Cereb Blood Flow Metab. 2005;25:1634–1644. doi: 10.1038/sj.jcbfm.9600160.
110.
Dabbagh K, Lewis DB. Toll-like receptors and T-helper-1/T-helper-2 responses. Curr Opin Infect Dis. 2003;16:199–204. doi: 10.1097/01.qco.0000073767.11390.47.
111.
Becker KJ, Kalil AJ, Tanzi P, Zierath DK, Savos AV, Gee JM, Hadwin J, Carter KT, Shibata D, Cain KC. Autoimmune responses to the brain after stroke are associated with worse outcome. Stroke. 2011;42:2763–2769. doi: 10.1161/STROKEAHA.111.619593.
112.
Mann DL. Innate immunity and the failing heart: the cytokine hypothesis revisited. Circ Res. 2015;116:1254–1268. doi: 10.1161/CIRCRESAHA.116.302317.
113.
Rustenhoven J, Aalderink M, Scotter EL, Oldfield RL, Bergin PS, Mee EW, Graham ES, Faull RL, Curtis MA, Park TI, Dragunow M. TGF-beta1 regulates human brain pericyte inflammatory processes involved in neurovasculature function. J Neuroinflammation. 2016;13:37. doi: 10.1186/s12974-016-0503-0.
114.
Viereck J, Bang C, Foinquinos A, Thum T. Regulatory RNAs and paracrine networks in the heart. Cardiovasc Res. 2014;102:290–301. doi: 10.1093/cvr/cvu039.
115.
Vahidy FS, Parsha KN, Rahbar MH, Lee M, Bui TT, Nguyen C, Barreto AD, Bambhroliya AB, Sahota P, Yang B, Aronowski J, Savitz SI. Acute splenic responses in patients with ischemic stroke and intracerebral hemorrhage. J Cereb Blood Flow Metab. 2016;36:1012–1021. doi: 10.1177/0271678X15607880.
116.
Yoshimoto Y, Tanaka Y, Hoya K. Acute systemic inflammatory response syndrome in subarachnoid hemorrhage. Stroke. 2001;32:1989–1993.
117.
Dhar R, Diringer MN. The burden of the systemic inflammatory response predicts vasospasm and outcome after subarachnoid hemorrhage. Neurocrit Care. 2008;8:404–412. doi: 10.1007/s12028-008-9054-2.
118.
Yang L, Kong Y, Ren H, Li M, Wei CJ, Shi E, Jin WN, Hao J, Vandenbark AA, Offner H. Upregulation of cd74 and its potential association with disease severity in subjects with ischemic stroke. Neurochem Int. 2017;107:148–155. doi: 10.1016/j.neuint.2016.11.007.
119.
Tsujioka H, Imanishi T, Ikejima H, et al. 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. doi: 10.1016/j.jacc.2009.04.021.
120.
Winklewski PJ, Radkowski M, Demkow U. Cross-talk between the inflammatory response, sympathetic activation and pulmonary infection in the ischemic stroke. J Neuroinflammation. 2014;11:213. doi: 10.1186/s12974-014-0213-4.
121.
van der Bilt IA, 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. doi: 10.1016/j.jcrc.2015.12.005.
122.
Johnson JD, Campisi J, Sharkey CM, Kennedy SL, Nickerson M, Greenwood BN, Fleshner M. Catecholamines mediate stress-induced increases in peripheral and central inflammatory cytokines. Neuroscience. 2005;135:1295–1307. doi: 10.1016/j.neuroscience.2005.06.090.
123.
Tracey KJ. The inflammatory reflex. Nature. 2002;420:853–859. doi: 10.1038/nature01321.
124.
Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, Eaton JW, Tracey KJ. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–462. doi: 10.1038/35013070.
125.
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci. 2015;9:392. doi: 10.3389/fncel.2015.00392.
126.
Nagatomo Y, Tang WH. Intersections between microbiome and heart failure: revisiting the gut hypothesis. J Card Fail. 2015;21:973–980. doi: 10.1016/j.cardfail.2015.09.017.
127.
Adrie C, Parlato M, Salmi L, Adib-Conquy M, Bical O, Deleuze P, Fitting C, Cavaillon JM, Monchi M. Bacterial translocation and plasma cytokines during transcatheter and open-heart aortic valve implantation. Shock. 2015;43:62–67. doi: 10.1097/SHK.0000000000000262.
128.
Sun J, Wang F, Ling Z, Yu X, Chen W, Li H, Jin J, Pang M, Zhang H, Yu J, Liu J. Clostridium butyricum attenuates cerebral ischemia/reperfusion injury in diabetic mice via modulation of gut microbiota. Brain Res. 2016;1642:180–188. doi: 10.1016/j.brainres.2016.03.042.
129.
Bercik P, Collins SM, Verdu EF. Microbes and the gut-brain axis. Neurogastroenterol Motil. 2012;24:405–413. doi: 10.1111/j.1365-2982.2012.01906.x.
130.
Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, Sita G, Racchumi G, Ling L, Pamer EG, Iadecola C, Anrather J. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016;22:516–523. doi: 10.1038/nm.4068.
131.
Singh V, Roth S, Llovera G, Sadler R, Garzetti D, Stecher B, Dichgans M, Liesz A. Microbiota dysbiosis controls the neuroinflammatory response after stroke. J Neurosci. 2016;36:7428–7440. doi: 10.1523/JNEUROSCI.1114-16.2016.
132.
Yin J, Liao SX, He Y, Wang S, Xia GH, Liu FT, Zhu JJ, You C, Chen Q, Zhou L, Pan SY, Zhou HW. Dysbiosis of gut microbiota with reduced trimethylamine-n-oxide level in patients with large-artery atherosclerotic stroke or transient ischemic attack. J Am Heart Assoc. 2015;4:1–12. doi: 10.1161/JAHA.115.002699.
133.
Winek K, Engel O, Koduah P, Heimesaat MM, Fischer A, Bereswill S, Dames C, Kershaw O, Gruber AD, Curato C, Oyama N, Meisel C, Meisel A, Dirnagl U. Depletion of cultivatable gut microbiota by broad- spectrum antibiotic pretreatment worsens outcome after murine stroke. Stroke. 2016;47:1354–1363. doi: 10.1161/STROKEAHA.115.011800.
134.
Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922.
135.
Crapser J, Ritzel R, Verma R, Venna VR, Liu F, Chauhan A, Koellhoffer E, Patel A, Ricker A, Maas K, Graf J, McCullough LD. Ischemic stroke induces gut permeability and enhances bacterial translocation leading to sepsis in aged mice. Aging (Albany NY). 2016;8:1049–1063. doi: 10.18632/aging.100952.
136.
Yamashiro K, Tanaka R, Urabe T, Ueno Y, Yamashiro Y, Nomoto K, Takahashi T, Tsuji H, Asahara T, Hattori N. Gut dysbiosis is associated with metabolism and systemic inflammation in patients with ischemic stroke. PLoS One. 2017;12:e0171521. doi: 10.1371/journal.pone.0171521.
137.
Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–455. doi: 10.1038/nature12726.
138.
Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165:111–124. doi: 10.1016/j.cell.2016.02.011.
139.
Li XS, Obeid S, Klingenberg R, et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J. 2017;38:814–824. doi: 10.1093/eurheartj/ehw582.
140.
Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–383. doi: 10.1083/jcb.201211138.
141.
Blann A, Shantsila E, Shantsila A. Microparticles and arterial disease. Semin Thromb Hemost. 2009;35:488–496. doi: 10.1055/s-0029-1234144.
142.
Martinez MC, Tual-Chalot S, Leonetti D, Andriantsitohaina R. Microparticles: targets and tools in cardiovascular disease. Trends Pharmacol Sci. 2011;32:659–665. doi: 10.1016/j.tips.2011.06.005.
143.
Lackner P, Dietmann A, Beer R, Fischer M, Broessner G, Helbok R, Marxgut J, Pfausler B, Schmutzhard E. Cellular microparticles as a marker for cerebral vasospasm in spontaneous subarachnoid hemorrhage. Stroke. 2010;41:2353–2357. doi: 10.1161/STROKEAHA.110.584995.
144.
Huang M, Hu YY, Dong XQ. High concentrations of procoagulant microparticles in the cerebrospinal fluid and peripheral blood of patients with acute basal ganglia hemorrhage are associated with poor outcome. Surg Neurol. 2009;72:481–489; discussion 489. doi: 10.1016/j.surneu.2008.12.016.
145.
Mesri M, Altieri DC. Leukocyte microparticles stimulate endothelial cell cytokine release and tissue factor induction in a JNK1 signaling pathway. J Biol Chem. 1999;274:23111–23118.
146.
Schoch B, Regel JP, Wichert M, Gasser T, Volbracht L, Stolke D. Analysis of intrathecal interleukin-6 as a potential predictive factor for vasospasm in subarachnoid hemorrhage. Neurosurgery. 2007;60:828–836; discussion 828. doi: 10.1227/01.NEU.0000255440.21495.80.
147.
Amabile N, Guérin AP, Leroyer A, Mallat Z, Nguyen C, Boddaert J, London GM, Tedgui A, Boulanger CM. Circulating endothelial microparticles are associated with vascular dysfunction in patients with end-stage renal failure. J Am Soc Nephrol. 2005;16:3381–3388. doi: 10.1681/ASN.2005050535.
148.
Agouni A, Lagrue-Lak-Hal AH, Ducluzeau PH, Mostefai HA, Draunet-Busson C, Leftheriotis G, Heymes C, Martinez MC, Andriantsitohaina R. Endothelial dysfunction caused by circulating microparticles from patients with metabolic syndrome. Am J Pathol. 2008;173:1210–1219. doi: 10.2353/ajpath.2008.080228.
149.
Boulanger CM, Scoazec A, Ebrahimian T, Henry P, Mathieu E, Tedgui A, Mallat Z. Circulating microparticles from patients with myocardial infarction cause endothelial dysfunction. Circulation. 2001;104:2649–2652.
150.
Sanborn MR, Thom SR, Bohman LE, Stein SC, Levine JM, Milovanova T, Maloney-Wilensky E, Frangos S, Kumar MA. Temporal dynamics of microparticle elevation following subarachnoid hemorrhage. J Neurosurg. 2012;117:579–586. doi: 10.3171/2012.6.JNS111163.
151.
Boettinger S, Lackner P. Cellular microparticles in subarachnoid hemorrhage. Transl Stroke Res. 2015;6:342–344. doi: 10.1007/s12975-015-0413-y.
152.
Sinauridze EI, Kireev DA, Popenko NY, Pichugin AV, Panteleev MA, Krymskaya OV, Ataullakhanov FI. Platelet microparticle membranes have 50- to 100-fold higher specific procoagulant activity than activated platelets. Thromb Haemost. 2007;97:425–434.
153.
Horn P, Erkilet G, Veulemans V, Kröpil P, Schurgers L, Zeus T, Heiss C, Kelm M, Westenfeld R. Microparticle- induced coagulation relates to coronary artery atherosclerosis in severe aortic valve stenosis. PLoS One. 2016;11:e0151499. doi: 10.1371/journal.pone.0151499.
154.
Varon D, Haion Y, Brill A, Leker R. Cell-driven angiogenesis and neurogenesis after stroke is regulated by platelet’s microparticles. Blood. 2008;112:1899–1899.
155.
Porro C, Trotta T, Panaro MA. Microvesicles in the brain: biomarker, messenger or mediator? J Neuroimmunol. 2015;288:70–78. doi: 10.1016/j.jneuroim.2015.09.006.
156.
Bianco F, Pravettoni E, Colombo A, Schenk U, Möller T, Matteoli M, Verderio C. Astrocyte-derived ATP induces vesicle shedding and IL-1 beta release from microglia. J Immunol. 2005;174:7268–7277.
157.
Bianco F, Perrotta C, Novellino L, Francolini M, Riganti L, Menna E, Saglietti L, Schuchman EH, Furlan R, Clementi E, Matteoli M, Verderio C. Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO J. 2009;28:1043–1054. doi: 10.1038/emboj.2009.45.
158.
Tian Y, Salsbery B, Wang M, Yuan H, Yang J, Zhao Z, Wu X, Zhang Y, Konkle BA, Thiagarajan P, Li M, Zhang J, Dong JF. Brain-derived microparticles induce systemic coagulation in a murine model of traumatic brain injury. Blood. 2015;125:2151–2159. doi: 10.1182/blood-2014-09-598805.
159.
Zhao Z, Wang M, Tian Y, Hilton T, Salsbery B, Zhou EZ, Wu X, Thiagarajan P, Boilard E, Li M, Zhang J, Dong JF. Cardiolipin-mediated procoagulant activity of mitochondria contributes to traumatic brain injury-associated coagulopathy in mice. Blood. 2016;127:2763–2772. doi: 10.1182/blood-2015-12-688838.
160.
Chen J, Venkat P, Zacharek A, Chopp M. Neurorestorative therapy for stroke. Front Hum Neurosci. 2014;8:382. doi: 10.3389/fnhum.2014.00382.
161.
Landgraf P, Rusu M, Sheridan R, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129:1401–1414. doi: 10.1016/j.cell.2007.04.040.
162.
Tan KS, Armugam A, Sepramaniam S, Lim KY, Setyowati KD, Wang CW, Jeyaseelan K. Expression profile of MicroRNAs in young stroke patients. PLoS One. 2009;4:e7689. doi: 10.1371/journal.pone.0007689.
163.
Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15:261–271. doi: 10.1016/j.devcel.2008.07.002.
164.
Long G, Wang F, Li H, Yin Z, Sandip C, Lou Y, Wang Y, Chen C, Wang DW. Circulating miR-30a, miR-126 and let-7b as biomarker for ischemic stroke in humans. BMC Neurol. 2013;13:178. doi: 10.1186/1471-2377-13-178.
165.
Qiang L, Hong L, Ningfu W, Huaihong C, Jing W. Expression of miR-126 and miR-508-5p in endothelial progenitor cells is associated with the prognosis of chronic heart failure patients. Int J Cardiol. 2013;168:2082–2088. doi: 10.1016/j.ijcard.2013.01.160.
166.
Long G, Wang F, Duan Q, Chen F, Yang S, Gong W, Wang Y, Chen C, Wang DW. Human circulating microRNA-1 and microRNA-126 as potential novel indicators for acute myocardial infarction. Int J Biol Sci. 2012;8:811–818. doi: 10.7150/ijbs.4439.
167.
De Rosa S, Fichtlscherer S, Lehmann R, Assmus B, Dimmeler S, Zeiher AM. Transcoronary concentration gradients of circulating microRNAs. Circulation. 2011;124:1936–1944. doi: 10.1161/CIRCULATIONAHA.111.037572.
168.
Chen J, Cui C, Yang X, Xu J, Venkat P, Zacharek A, Yu P, Chopp M. Mir-126 affects brain-heart interaction after cerebral ischemic stroke [published online ahead of print January 19, 2017]. Transl Stroke Res. doi: 10.1007/s12975-017-0520-z.
169.
Dharap A, Bowen K, Place R, Li LC, Vemuganti R. Transient focal ischemia induces extensive temporal changes in rat cerebral microRNAome. J Cereb Blood Flow Metab. 2009;29:675–687. doi: 10.1038/jcbfm.2008.157.
170.
Climent M, Quintavalle M, Miragoli M, Chen J, Condorelli G, Elia L. TGFβ triggers miR-143/145 transfer from smooth muscle cells to endothelial cells, thereby modulating vessel stabilization. Circ Res. 2015;116:1753–1764. doi: 10.1161/CIRCRESAHA.116.305178.
171.
Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, Weber M, Hamm CW, Röxe T, Müller-Ardogan M, Bonauer A, Zeiher AM, Dimmeler S. Circulating microRNAs in patients with coronary artery disease. Circ Res. 2010;107:677–684. doi: 10.1161/CIRCRESAHA.109.215566.
172.
van der Bilt I, Hasan D, van den Brink R, et al.; SEASAH (Serial Echocardiography After Subarachnoid Hemorrhage) Investigators. Cardiac dysfunction after aneurysmal subarachnoid hemorrhage: relationship with outcome. Neurology. 2014;82:351–358. doi: 10.1212/WNL.0000000000000057.
173.
Tanabe M, Crago EA, Suffoletto MS, Hravnak M, Frangiskakis JM, Kassam AB, Horowitz MB, Gorcsan J. Relation of elevation in cardiac troponin I to clinical severity, cardiac dysfunction, and pulmonary congestion in patients with subarachnoid hemorrhage. Am J Cardiol. 2008;102:1545–1550. doi: 10.1016/j.amjcard.2008.07.053.
174.
Wybraniec MT, Mizia-Stec K, Krzych Ł. Neurocardiogenic injury in subarachnoid hemorrhage: a wide spectrum of catecholamin-mediated brain-heart interactions. Cardiol J. 2014;21:220–228. doi: 10.5603/CJ.a2014.0019.
175.
Seo JY, Lee KB, Lee JG, Kim JS, Roh H, Ahn MY, Park BW, Hyon MS. Implication of left ventricular diastolic dysfunction in cryptogenic ischemic stroke. Stroke. 2014;45:2757–2761. doi: 10.1161/STROKEAHA.114.006108.
176.
Christensen H, Fogh Christensen A, Boysen G. Abnormalities on ECG and telemetry predict stroke outcome at 3 months. J Neurol Sci. 2005;234:99–103. doi: 10.1016/j.jns.2005.03.039.
177.
Lane RD, Wallace JD, Petrosky PP, Schwartz GE, Gradman AH. Supraventricular tachycardia in patients with right hemisphere strokes. Stroke. 1992;23:362–366.
178.
Naidech AM, Kreiter KT, Janjua N, Ostapkovich ND, Parra A, Commichau C, Fitzsimmons BF, Connolly ES, Mayer SA. Cardiac troponin elevation, cardiovascular morbidity, and outcome after subarachnoid hemorrhage. Circulation. 2005;112:2851–2856. doi: 10.1161/CIRCULATIONAHA.105.533620.
179.
Banki N, Kopelnik A, Tung P, Lawton MT, Gress D, Drew B, Dae M, Foster E, Parmley W, Zaroff J. Prospective analysis of prevalence, distribution, and rate of recovery of left ventricular systolic dysfunction in patients with subarachnoid hemorrhage. J Neurosurg. 2006;105:15–20. doi: 10.3171/jns.2006.105.1.15.
180.
Ahtarovski KA, Iversen KK, Christensen TE, Andersson H, Grande P, Holmvang L, Bang L, Hasbak P, Lønborg JT, Madsen PL, Engstrøm T, Vejlstrup NG. Takotsubo cardiomyopathy, a two-stage recovery of left ventricular systolic and diastolic function as determined by cardiac magnetic resonance imaging. Eur Heart J Cardiovasc Imaging. 2014;15:855–862. doi: 10.1093/ehjci/jeu004.
181.
Mayer SA, Lin J, Homma S, Solomon RA, Lennihan L, Sherman D, Fink ME, Beckford A, Klebanoff LM. Myocardial injury and left ventricular performance after subarachnoid hemorrhage. Stroke. 1999;30:780–786.
182.
Oras J, Grivans C, Dalla K, Omerovic E, Rydenhag B, Ricksten SE, Seeman-Lodding H. High- sensitive troponin T and N-terminal pro B-type natriuretic peptide for early detection of stress-induced cardiomyopathy in patients with subarachnoid hemorrhage. Neurocrit Care. 2015;23:233–242. doi: 10.1007/s12028-015-0108-y.
183.
Sandhu R, Aronow WS, Rajdev A, Sukhija R, Amin H, D’aquila K, Sangha A. Relation of cardiac troponin I levels with in-hospital mortality in patients with ischemic stroke, intracerebral hemorrhage, and subarachnoid hemorrhage. Am J Cardiol. 2008;102:632–634. doi: 10.1016/j.amjcard.2008.04.036.
184.
Rost NS, Wolf PA, Kase CS, Kelly-Hayes M, Silbershatz H, Massaro JM, D’Agostino RB, Franzblau C, Wilson PW. Plasma concentration of C-reactive protein and risk of ischemic stroke and transient ischemic attack: the Framingham study. Stroke. 2001;32:2575–2579.
185.
Di Napoli M, Papa F, Bocola V. C-reactive protein in ischemic stroke: an independent prognostic factor. Stroke. 2001;32:917–924.
186.
Ridker PM, Glynn RJ, Hennekens CH. C-reactive protein adds to the predictive value of total and HDL cholesterol in determining risk of first myocardial infarction. Circulation. 1998;97:2007–2011.

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research

Visualization of Hypoxia-Induced Endoplasmic Reticulum Stress Regulating lncRNA (HypERlnc) using RNA fluorescent in situ hybridization. Nucleus in blue, HypERlnc in red. See related article, page 368.

Circulation Research
Pages: 451 - 468
PubMed: 28775014

History

Published online: 4 August 2017
Published in print: 4 August 2017

Permissions

Request permissions for this article.

Keywords

  1. brain injury
  2. brain ischemia
  3. heart failure
  4. inflammation
  5. stroke

Subjects

Authors

Affiliations

Zhili Chen*
From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
Poornima Venkat*
From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
Don Seyfried*
From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
Michael Chopp
From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
Tao Yan
From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
Jieli Chen
From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).

Notes

*
These authors contributed equally to this article.
Correspondence to Jieli Chen, MD, Gerontology and Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China. E-mail [email protected]; or Tao Yan, MD, PhD, Gerontology and Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China. E-mail [email protected]

Disclosures

None.

Sources of Funding

This work was supported by the National Institute of Neurological Disorders and Stroke R01 NS083078-01A1 (Dr Chen), R01 NS099030-01 (Dr Chen), and R01NS097747 (Dr Chen); by the American Heart Association 17POST33410580 (Dr Venkat); and by the National Natural Science Foundation of China grant 81300993 and 81571145 (Dr Yan).

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Association of the blood urea nitrogen to serum albumin ratio and all-cause mortality in critical ill acute ischemic stroke patients: a retrospective cohort study of MIMIC-IV database 3.0, Frontiers in Nutrition, 11, (2025).https://doi.org/10.3389/fnut.2024.1509284
    Crossref
  2. Nonlinear Relationship Between Blood Urea Nitrogen to Albumin Ratio and 3-Month Outcomes in Patients With Acute Ischemic Stroke: A Second Analysis Based on a Prospective Cohort Study, World Neurosurgery, 194, (123452), (2025).https://doi.org/10.1016/j.wneu.2024.11.035
    Crossref
  3. Emerging advances in drug delivery systems (DDSs) for optimizing cancer complications, Materials Today Bio, 30, (101375), (2025).https://doi.org/10.1016/j.mtbio.2024.101375
    Crossref
  4. Prevalence of neurogenic stress cardiomyopathy in acute ischemic stroke and relationship with leukocytosis, Journal of Stroke and Cerebrovascular Diseases, 34, 1, (108169), (2025).https://doi.org/10.1016/j.jstrokecerebrovasdis.2024.108169
    Crossref
  5. Naoxintong capsule attenuates heart damage after ischemic stroke via Nuclear factor-κB / Pyrin domain-containing protein 3 / Caspase-1 signaling, Journal of Ethnopharmacology, 341, (119240), (2025).https://doi.org/10.1016/j.jep.2024.119240
    Crossref
  6. Circulating mitochondria carrying cGAS promote endothelial Secreted group IIA phospholipase A2-mediated neuroinflammation through activating astroglial/microglial Integrin-alphavbeta3 in subfornical organ to augment central sympathetic overdrive in heart failure rats, International Immunopharmacology, 144, (113649), (2025).https://doi.org/10.1016/j.intimp.2024.113649
    Crossref
  7. Causal associations between hypertension and abnormal brain cortical structures: Insights from a bidirectional Mendelian randomization study, International Journal of Cardiology Cardiovascular Risk and Prevention, 24, (200354), (2025).https://doi.org/10.1016/j.ijcrp.2024.200354
    Crossref
  8. Navigating the Statin Landscape: A Comprehensive Review of Stroke Prevention Strategies, Cureus, (2024).https://doi.org/10.7759/cureus.53555
    Crossref
  9. The Clock is Ticking: Overcoming Time with Mechanical Thrombectomy for Middle Cerebral Artery Infarct, European Journal of Therapeutics, 30, 3, (368-371), (2024).https://doi.org/10.58600/eurjther2076
    Crossref
  10. Utilizing engineered extracellular vesicles as delivery vectors in the management of ischemic stroke: a special outlook on mitochondrial delivery, Neural Regeneration Research, 20, 8, (2181-2198), (2024).https://doi.org/10.4103/NRR.NRR-D-24-00243
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Brain–Heart Interaction
Circulation Research
  • Vol. 121
  • No. 4

Purchase access to this journal for 24 hours

Circulation Research
  • Vol. 121
  • No. 4
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response