Next Article in Journal
Structural Heart Alterations in Brugada Syndrome: Is it Really a Channelopathy? A Systematic Review
Previous Article in Journal
Effect of Fetoscopic Laser Photocoagulation on Fetal Growth and Placental Perfusion in Twin–Twin Transfusion Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Mental Stress on Cardiovascular Health—Part II

1
Institute of Public Health and Clinical Medicine, Umea University, 90187 Umea, Sweden
2
Brunel University, Middlesex, London UB8 3PH, UK
3
St. George’s University, London SW17 0RE, UK
4
Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy
5
Cardiovascular and Interventional Department, S. Elia Hospital, 93100 Caltanissetta, Italy
6
Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(15), 4405; https://doi.org/10.3390/jcm11154405
Submission received: 16 June 2022 / Revised: 23 July 2022 / Accepted: 26 July 2022 / Published: 28 July 2022
(This article belongs to the Section Cardiovascular Medicine)

Abstract

:
Endothelial dysfunction is one of the earliest manifestations of atherosclerosis, contributing to its development and progression. Mental stress induces endothelial dysfunction through increased activity of the sympathetic nervous system, release of corticotropin-releasing hormone from the hypothalamus, inhibition of nitric oxide (NO) synthesis by cortisol, and increased levels of pro-inflammatory cytokines. Mental-stress-induced increased output of the sympathetic nervous system and concomitant withdrawal of the parasympathetic inflammatory reflex results in systemic inflammation and activation of a neural–hematopoietic–arterial axis. This includes the brainstem and subcortical regions network, bone marrow activation, release of leukocytes into the circulation and their migration to the arterial wall and atherosclerotic plaques. Low-grade, sterile inflammation is involved in all steps of atherogenesis, from coronary plaque formation to destabilisation and rupture. Increased sympathetic tone may cause arterial smooth-muscle-cell proliferation, resulting in vascular hypertrophy, thus contributing to the development of hypertension. Emotional events also cause instability of cardiac repolarisation due to brain lateralised imbalance of cardiac autonomic nervous stimulation, which may lead to asymmetric repolarisation and arrhythmia. Acute emotional stress can also provoke severe catecholamine release, leading to direct myocyte injury due to calcium overload, known as myocytolysis, coronary microvascular vasoconstriction, and an increase in left ventricular afterload. These changes can trigger a heart failure syndrome mimicking acute myocardial infarction, characterised by transient left ventricular dysfunction and apical ballooning, known as stress (Takotsubo) cardiomyopathy. Women are more prone than men to develop mental-stress-induced myocardial ischemia (MSIMI), probably reflecting gender differences in brain activation patterns during mental stress. Although guidelines on CV prevention recognise psychosocial factors as risk modifiers to improve risk prediction and decision making, the evidence that their assessment and treatment will prevent CAD needs further evaluation.

1. Introduction

Epidemiological studies suggest that acute and chronic mental stress, such as anger, fear, job strain, depression or exposure to natural disasters, may contribute to the pathogenesis and development of coronary artery disease (CAD) [1,2,3,4]. The first part of the review on the relationship between psychological stress and cardiovascular (CV) disease focused on the brain response to negative emotions and autonomic nervous system imbalance, leading to hemodynamic and coronary artery responses to mental stress [5]. The aim of Part II is to review the evidence for the clinical association between mental stress and systemic CV effects, including endothelial dysfunction, inflammation, hypertension, arrhythmia, and stress cardiomyopathy, based on most recent literature findings.

2. Recognised Underlying Mechanisms That Can Be Measured in Laboratory

Effects of mental stress on vascular endothelium. The functional integrity of the vascular endothelium has a critical role in transducing mental stress into coronary artery disease (CAD). The endothelium lines the vascular inner wall and regulates homeostatic functions such as vascular tone, permeability, proliferation of vascular smooth muscle cells, platelet aggregation, and balance between thrombogenesis and fibrinolysis [6,7,8]. It is the main factor regulating coronary blood flow. Since myocardial oxygen extraction is almost maximal at rest, increases in myocardial metabolic activity must be balanced by proportional increases in coronary blood flow. Flow resistance is inversely proportional to the fourth power of the radius; hence, the distribution of coronary blood flow is strictly dependent on the arterial diameter. Under resting conditions, the endothelium maintains high vascular tone [9]. Under physiological stimuli, such as exercise, the endothelial cells modulate an appropriate dilatation of coronary arteries by locally releasing nitric oxide (NO), thus allowing the increase in coronary blood flow [10]. NO also has anti-inflammatory properties which protect the integrity of the endothelium by inhibiting fibrosis, platelet aggregation, and apoptosis [11,12].
The endothelium releases endothelium-derived contracting factor, endothelin-1 (ET-1), which mediates the vasoconstriction response to noradrenaline, particularly in the microvascular bed, balancing vasodilation with vasoconstriction. Clinical studies have shown that public speaking and anger provocation are associated with increased endothelial-cell-derived microparticles, which derive from the membranes of apoptotic endothelial cells and serve as a marker of endothelial damage [13,14].
Endothelial dysfunction is characterised by reduced bioavailability of NO and increased endothelin, leading to an imbalance between vasodilatation and vasoconstriction. Endothelial dysfunction can be identified by measuring flow-mediated dilatation (FMD). During brachial FMD testing, endothelium-dependent vasodilatation is induced by reactive hyperaemia, due to an increase in endothelial shear stress [15]. The underlying mechanisms for mental-stress-induced endothelial dysfunction include (a) increased activity of the sympathetic nervous system, which induces endothelial dysfunction by direct stimulation of α-adrenergic receptors, through noradrenaline released in the arterial walls; (b) the release of corticotropin-releasing hormone from the hypothalamus, which occurs within seconds following exposure to stress, contributing to endothelial dysfunction by stimulating the release of the vasoconstrictor ET-1 from the endothelium; (c) release of cortisol, 20–40 min after the onset of stress, which impairs endothelial function by inhibiting NO synthesis and increasing ET-1 release; (d) increased levels of pro-inflammatory cytokines, such as interleukins and tumour necrosis factor alpha, that directly impair endothelial function [11,14,16,17,18]. Additionally, mental stress may act in combination with the conventional cardiovascular risk factors to accelerate the development of atherosclerosis [19]. This may explain the limited effect of negative emotions on atherosclerosis in young age.
The immediate response to acute mental stress depends on the activity of the sympathetic nervous system, while the delayed response is regulated by release of cortisol and ET-1 [16]. In healthy subjects without clinical evidence of CAD, a brief episode of mental stress can induce a rapid impairment of endothelium-dependent vasodilatation that may last for hours, even after resolution of the hemodynamic changes [16,20]. This suggests that transient, repeated stress, as encountered during daily life, may result in clinically relevant effects, such as atherosclerosis and MSIMI [21]. Moreover, individuals with clinical depression have impaired endothelial function, which may account for their increased risk of CAD [22].
Endothelial dysfunction is one of the earliest manifestations of atherosclerosis, often preceding angiographic evidence of atherosclerotic plaque or increased intima-media ratio on ultrasound examination, contributing to its development and progression [23,24]. In healthy endothelium, dilatation occurs in response to changes in localised blood flow, shear stress or increased resistance. Impairment of endothelial control of vascular tone in small resistance arteries, which are responsible for most of the total peripheral resistance, can greatly affect tissue perfusion and blood pressure, and can be critical for myocardial perfusion. In addition, endothelial dysfunction induces activation of macrophages, proliferation of vascular smooth muscle cells, and platelet aggregation and adhesion to the walls of blood vessels, leading to both chronic stable myocardial ischemia and acute ischemic events [10].
Impairment of endothelium-derived vasodilatation may simultaneously occur in coronary microcirculation, epicardial arteries, and peripheral arterial systems, thus representing a systemic arterial disorder [24,25]. However, a segmental pattern along the coronary arteries has also been demonstrated [26]. In these patients, coronary segments with abnormal endothelial function are associated with lipid-rich plaques and necrotic core areas, which contain sites of active inflammation, microcalcifications and expression of vulnerable plaque. Inflammation may be a link between endothelial dysfunction and plaque composition [27,28]. Serial intravascular ultrasound evaluations have found accelerated progression of atherosclerosis in coronary segments with endothelial dysfunction [29].
Inflammatory response to mental stress. Mental-stress-induced sympathetic activation drives vascular inflammation and promotes progression of atherosclerosis, plaque destabilisation, and increased risk of developing heart failure [30,31,32,33,34]. Systemic inflammation is involved in all steps of atherogenesis, from plaque formation to rupture [35,36]. Chronic stress may contribute to atherosclerosis by inducing systemic, sterile, low-grade inflammation, different from the response to acute infections [37]. Some imaging studies have shown that the activity of the amygdala is increased under conditions of both acute or chronic stress [34,38,39]. The amygdala regulates the stress perception and emotional response of the central nervous system network associated with carotid and coronary arterial inflammation, and hence it is implicated in atherosclerosis progression and plaque destabilisation [40,41].
In normal conditions the vagal (parasympathetic) efferent innervations release acetylcholine both in the reticuloendothelial system and in the heart, blocking the release of inflammatory cytokines, including interleukins (IL-1, IL-2, IL-6) and tumour necrosis factor alpha (TNF-α), thus modulating the inflammatory response to acute inflammation. This cholinergic anti-inflammatory pathway is also termed “parasympathetic inflammatory reflex” [42,43,44,45]. Mental stress induces an imbalance in autonomic regulation, resulting in increased output of the sympathetic nervous system and concomitant parasympathetic (vagal) nervous system withdrawal. Thus, reduced parasympathetic activity with mental stress results in the release of inflammatory cytokines by immunocompetent cells expressing acetylcholine receptors, including myocardial vasculature macrophages [46,47,48]. In turn, TNF-α can provoke the release of the vasoconstrictor ET-1 from activated macrophages, thereby promoting vasoconstriction and endothelial dysfunction, leading to coronary plaque rupture with clinical consequences [49]. The inflammatory response is also characterised by increased levels of C-reactive protein (CRP), an acute phase reactant used as a sensitive marker of systemic inflammation, strongly associated with the risk of CAD, ischemic stroke, and vascular mortality [50,51,52,53].
Mental stress also promotes the activation of a neural–hematopoietic–arterial axis, which includes the brain stress network, bone marrow, and arterial inflammation (Figure 1) [38,54,55]. The mental-stress-induced release of noradrenaline promotes hematopoietic stem cell proliferation in the bone marrow, leading to an increased release of leukocytes, in particular neutrophils and monocytes, into the circulation [56]. In conditions of chronic inflammation, high catecholamine levels induce pro-inflammatory changes in monocytes [57]. These inflammatory cells migrate to the arterial wall and infiltrate the atherosclerotic plaques, where they induce inflammation [58]. Proteases released from inflammatory leukocytes weaken the plaque fibrous cap, which may lead to plaque rupture and acute coronary events [59,60,61]. The importance of bone marrow activation and arterial inflammation in the development of atherosclerosis is confirmed by the observation that higher blood levels of monocytes and neutrophils correlate with increased CAD incidence and mortality [62].
Inflammatory mediators induced by chronic stress, in particular IL-6, activate the HPA axis, which is involved in the containment of inflammatory reactions, resulting in cortisol secretion by the adrenal gland [63]. However, the mechanisms by which cortisol influences atherosclerosis are not well defined. Although cortisol has anti-inflammatory actions, chronic exposure to elevated levels may induce a state of resistance, reducing the anti-inflammatory actions while increasing the concentration of inflammatory cytokines [64]. This effect may account for the association between increased circulating cortisol levels and increased perivascular inflammation, endothelial dysfunction, and progression of coronary artery calcification [18,65,66]. However, prolonged activation of the HPA axis due to chronic stress or repeated episodes of acute stress may lead to its dysfunction, resulting in reduced anti-inflammatory activity of cortisol and progression of the inflammatory response in the arterial wall [67,68].
Mental stress activation of the sympathetic nervous system also induces platelet activation. Activated platelets release inflammatory cytokines and adhesive molecules, which induce leukocyte recruitment on the endothelial surface, resulting in increased coagulability and a prothrombotic state. In turn, these conditions enhance the recruitment of leukocytes and platelets on the vessel wall, promoting the development of arterial thrombosis [31,69].
The central role of inflammation in the pathogenesis and clinical manifestation of atherosclerosis has also been confirmed by the reduction in coronary events by colchicine, an anti-inflammatory drug that reduces the inflammatory activity of leukocytes [70,71]. Moreover, in stable patients with previous myocardial infarction and persistent systemic inflammatory response, blocking the interleukin-6 activity directly using the monoclonal antibody canakinumab resulted in significant reduction in high-sensitivity CRP levels and incidence of subsequent CV events, without changes in low-density lipoprotein cholesterol [72].

3. Clinical Conditions Associated with Mental Stress

Hypertension. Epidemiological studies provide support for the importance of psychological stress in the development of essential hypertension, although this relationship is still debatable [73,74,75,76,77,78,79]. Mental-stress-induced autonomic imbalance, with increased sympathetic tone accompanied by reduced parasympathetic tone, may cause smooth-muscle-cell proliferation and vascular remodelling, resulting in vasoconstriction [80]. Cortisol stress reactivity, an index of HPA axis function, is another possible mechanism through which mental stress may influence the risk of hypertension [81]. Longitudinal studies have shown that greater cardiovascular (CV) responses to stressful tasks are associated with long-term development of hypertension and with increased risk of cardiovascular mortality, independently of conventional CV risk factors [82,83,84,85,86]. The repeated episodes of CV activation result in vascular hypertrophy, leading to a progressive increase in peripheral resistance and thus contributing to the development of established hypertension. Moreover, increased systolic blood pressure reactivity to mental stress is positively associated with progression of atherosclerosis, as expressed by carotid intima-media thickness (IMT), an early marker of generalised atherosclerosis, including coronary arteries, strongly associated with CAD [87,88].
Arrhythmia. Both acute and chronic stressful events increase the risk of arrhythmia and sudden cardiac death (SCD) [89,90,91]. Ventricular arrhythmias in patients fitted with an implantable cardioverter-defibrillator (ICD) are 5 times more frequent after heightened anger than in control periods [92]. Anger and negative emotions are also associated with the initiation and progression of atrial fibrillation after adjusting for conventional CV risk factors, previous CAD, and valvar heart disease [93,94,95]. In individuals with inherited arrhythmogenic disorders characterised by congenital myocardial ion channelopathies, such as long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT), emotional stress, as well as physical exercise, can induce potentially lethal ventricular tachyarrhythmias, in the absence of structural heart disease [96,97,98,99,100,101]. Cardiac autonomic dysfunction, characterised by enhanced sympathetic activity and parasympathetic withdrawal, is the link between mental stress and arrhythmia. Emotional factors may influence the risk of arrhythmia by inducing electric heterogeneities of ventricular depolarisation or repolarisation [102,103]. The myocardial ion channels are responsible for the transmembrane flow of sodium, potassium and calcium ions that determine the cardiac action potential and intracellular calcium homeostasis. In normal hearts, sympathetic activation shortens the action potential as the heart rate increases. In LQTS, a defective cardiac potassium channel results in a paradoxical increase in the QT interval due to pathological prolongation of the plateau phase of the action potential, leading to abnormal depolarisations during the repolarisation phase of the action potential, defined as early afterdepolarisations (EADs), which can cause lethal arrhythmias [104,105].
The genetic mutation in CPVT alters intracellular calcium handling. In addition to normal calcium release during systole, which underlies cardiac contraction, there is a spontaneous calcium release during diastole. As a consequence, sympathetic activation results in intracellular calcium overload, inducing oscillations of membrane potentials occurring after full repolarisation, known as delayed afterdepolarisations (DADs), which can trigger ventricular arrhythmias [106,107].
Beta-blockers (BB), especially non-selective nadolol and propranolol, are a first-line therapy for patients with a clinical diagnosis of LQTS or CPVT, with proven effects on arrhythmia and mortality reduction [108,109]. In patients with LQTS, in addition to BB, an implantable cardioverter-defibrillator (ICD) should be considered for those who experienced syncope or cardiac arrest. When BB and/or ICD are not effective or contraindicated, surgical left cardiac sympathetic denervation that reduces the catecholaminergic effect on left ventricular myocardium may be considered [96]. In patients with CPVT, in addition to BB and ICD, flecainide, alone or with BB, prevents ventricular arrhythmias. Patients experiencing episodes of multiple ventricular tachycardia/fibrillation over a short period of time (arrhythmic storm) may be treated with stellate ganglion blockade, using percutaneously infiltrating local anaesthetic agents [110].
Heterogeneity of repolarisation is also important in patients with heterogeneous cardiac denervation due to myocardial infarction or cardiomyopathies, inducing repolarisation instability, reflected in ECG T wave alternans, thereby increasing the risk of ventricular arrhythmia [91,111]. One suggested mechanism by which mental stress triggers arrhythmia is the “brain-heart laterality”, in which lateralised imbalance of autonomic neural stimulation of the heart may induce asymmetric repolarisation [112,113]. While the left sided sympathetic nerves are mainly distributed over the postero-inferior wall of the ventricles, the right sided are over the anterior wall, although with large interconnections. There is also anatomical evidence to support subcortical lateralisation of efferent sympathetic pathways, maintained from brainstem to peripheral nerves. Neuroimaging studies suggest that cortical regions in the right hemisphere, in particular right and ventral posterior insula subregions, exert sympathetic control [114,115]. Lateralisation of the processing of emotion in the brain and of autonomic inputs to the surface of the heart contribute to the repolarisation instability, which may lead to arrhythmia [116,117].
Takotsubo cardiomyopathy. Acute emotional stress may trigger a heart failure syndrome clinically presenting as acute ST elevation myocardial infarction, characterised by reversible left ventricular balloon-like wall motion abnormalities, usually involving the LV apex and rarely the mid ventricle, without significant coronary artery obstruction, termed Takotsubo syndrome (TTS) or “broken heart syndrome” (Figure 2 and Figure 3) [118,119,120]. The underlying cause is assumed to be an overstimulation of the sympathetic nervous system, triggered by emotional and physical stress, although, in a small proportion of patients, the syndrome is not directly related to any stressful event [121,122,123]. TTS shares common clinical presentation and pathophysiological mechanisms with “neurogenic stress cardiomyopathy”, which includes subarachnoid haemorrhage, stroke, either haemorrhagic or ischemic, traumatic brain injury, epilepsy, and central nervous system infections [124]. Depression and chronic anxiety disorders are frequent in TTS patients [125]. In addition to negative emotions, TTS can be rarely triggered by positive life events, including important personal achievements, marriage, and holidays. This condition has been described as “happy heart syndrome”. It is associated with clinical and laboratory findings similar to the “broken heart syndrome” [126,127]. The possible mechanism involved is central and subcortical brain areas, and the sympathetic activation induced by positive emotional excitement [128]. Despite being previously believed to be a transient benign disease, the syndrome carries high rates of complications, similar to negative outcomes of acute coronary events [122]. The higher prevalence of TTS in postmenopausal women seems to indicate that oestrogen deficiency may have a pathogenic role. Under physiological conditions, oestrogen has beneficial effects on coronary microcirculation tone and endothelial function, reducing the sympathetic response to mental stress and improving coronary blood flow [129,130,131]. During menopause, both increased sympathetic vasoconstriction and endothelial dysfunction are a consequence of reduced oestrogen levels [132].
Stress-induced cardiac sympathetic hyperactivity leads to an acute increase in myocardial and circulating catecholamine levels that induce direct myocyte injury likely due to calcium overload, coronary microvascular vasoconstriction, increased ventricular afterload resulting in oxygen supply–demand mismatch, endothelial dysfunction, and myocardial inflammation [121,133,134,135,136]. Histological alterations are typical of catecholamine toxicity, which consists of focal mononuclear inflammatory cells infiltration, a considerable increase in extracellular matrix protein levels, and contraction band necrosis (also known as myocytolysis). Neuroimaging studies have shown that patients with TTS have structural alterations in the limbic system and reduced functional connectivity among brain regions of the cortico-limbic network, comprising the prefrontal cortex, right insula, amygdala, hypothalamus, cingulate cortex, and thalamus, all of which regulate the autonomic nervous system and the CV response to mental stress [137,138,139,140,141]. The altered functional connectivity pattern may persist for weeks following the acute cardiac event, well after the recovery of left ventricular dysfunction [142]. Reduced functional brain connectivity may precede the development of TTS by years. A retrospective study of the alterations in neural connectivity in patients who subsequently developed TTS showed increased amygdalar activity years before developing TTS [143]. Such heightened amygdalar activity may predispose someone to TTS by potentiating the autonomic and neurohormonal response to future stressors.

4. Gender Differences in Response to Mental Stress

Women, especially young women, develop MSIMI more often than men [144,145,146]. There are gender differences in the brain activation pattern during mental stress [147,148]. Women with CAD have greater activation of cortico-limbic areas that regulate emotions and sympathetic response, including the amygdala, hippocampus, and prefrontal cortex, compared to men. This pattern of brain activation in women could translate into differences in neurohormonal and CV response to stress. Although men and women with CAD develop similar rate-pressure product response to mental stress, women have more marked endothelial dysfunction and intense vasoconstrictive response, resulting in microvascular dysfunction [144,149,150]. Microvascular dysfunction is common in women with chest pain, even in the absence of significant epicardial coronary obstruction [151]. This may be the substrate for MSIMI being more likely in women than in men.

5. Prevention and Treatment

Prevention strategies may be population-based (universal), involving the general population, or targeted to those at the highest risk of adverse events. Intervention studies of modifying stressors in healthy individuals are difficult to implement because they require changes in life habits, work organisation, and social support. In addition, stressors may be unpredictable and trigger CV events in very different conditions, such as family life, sport events and natural disasters. There are only few small general population randomised studies addressing this in the literature [152].
The rationale of focusing on high-CV-risk individuals is that given the low overall incidence rates of CV events in the healthy population, the reduction in absolute risk of CAD due to a reduction in stress-induced disease would be small. In contrast, in high-risk individuals, with high overall rates of CV events, the reduction in events due to stress would have larger effects [19]. In secondary prevention, cognitive behavioural therapy decreased the risk of recurrent CV events in patients with previous CAD and prolonged survival in women after acute myocardial infarction [153,154]. Treatment of depression in patients with recent myocardial infarction resulted in lower major coronary events compared to the placebo [155]. Treatment of major depression even in individuals without baseline CAD substantially reduced the development of coronary events [156]. In a small-size study of patients with stable CAD and MSIMI (see Part I [5]), treatment with escitalopram reduced the rate of MSIMI demonstrated on echocardiography and/or ECG, probably due to a beneficial effect on platelets aggregation [157]. However, other studies showed little or no evidence that treating depression in such patients improves CV outcomes [158,159]. A comprehensive systematic review of several studies of psychological treatments in patients with CAD, including those with and without a psychopathology at baseline, showed no effect on total mortality, myocardial revascularisation procedures and non-fatal myocardial infarction. However, there was low-quality evidence of a small reduction in cardiac mortality and improvement of psychological symptoms, such as depression, anxiety or stress [160]. Treatments have been more effective in patients with established psychological conditions at baseline. However, there was no impact on total mortality, risk of revascularisation procedures, or the rate of non-fatal myocardial infarction. In addition, most studies were of low quality, suggesting uncertainty about the results. Since exercise training is a crucial component of cardiac rehabilitation, and exercise has been shown to reduce depression symptoms in chronically ill patients, comprehensive cardiac rehabilitation including the psychological components should be recommended in patients with previous CAD [161,162]. Guidelines on CAD prevention recognise the relevance of psychosocial factors in the development of significant CAD. However, the evidence that their assessment and treatment may prevent CAD is still inconclusive [163,164].

6. Limitations

There are several limitations in the interpretation of the relationship between mental stress and CV disease. Firstly, the pathophysiological mechanisms linking mental stress to increased risk of CAD are complex and multifactorial, involving several variables, such as activation of brain circuits, the autonomic nervous system, the neuroendocrine and immune systems, conventional CV risk factors, and individual’s past experience. In turn, the resulting hemodynamic and inflammatory changes in their combined effects modulate the brain response to environmental stimuli [19,165]. These interactions may account for the large individual variability in the response to mental stress. This makes excluding confounding factors and identifying the role of mental stress in the development of CV disease difficult. Secondly, most available studies are observational and based on patient’s recalling of negative emotions preceding CV events. This method cannot determine whether the association between stress and CV event is causal [3]. Thirdly, there are large methodological and patient characteristic differences between studies [90]. Finally, some hemodynamic and vascular effects of mental stress may occur without symptoms or may develop long after the stressful event. Hence, the causal relationship cannot be precisely ascertained.

7. Conclusions

Over the past four decades, there has been an increasing awareness that mental stress is an important and potentially modifiable risk factor for acute and chronic CV events. Emerging epidemiological and experimental evidence suggest that stress-induced hemodynamic, vascular and inflammatory alterations may interact together and exert an important role in atherosclerosis progression, as well as in acute CV disease triggering, especially in high-risk individuals. However, in clinical practice, the assessment of the impact of mental stress on CAD disease is often missing. Despite current guidelines recognising the importance of mental stress as a CV risk factor, further research with large longitudinal studies is needed to define the prevention and treatment strategies required to reduce the CV effects of mental stress.

Author Contributions

Conceptualization, F.V. and M.Y.H.; writing original draft preparation, G.L. and S.V.; writing review and editing, M.Y.H. and F.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yusuf, S.; Hawken, S.; Ôunpuu, S.; Dans, T.; Avezum, A.; Lanas, F.; McQueen, M.; Budaj, A.; Pais, P.; Varigos, J.; et al. Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): Case-Control study. Lancet 2004, 364, 937–952. [Google Scholar] [CrossRef]
  2. Rosengren, A.; Hawken, S.; Ôunpuu, S.; Sliwa, K.; Zubaid, M.; Almahmeed, W.A.; Blackett, K.N.; Sitthi-amorn, C.; Sato, H.; Yusuf, S. Association of psychosocial risk factors with risk of acute myocardial infarction in 11,119 cases and 13,648 controls from 52 countries (the INTERHEART study): Case-Control study. Lancet 2004, 364, 953–962. [Google Scholar] [CrossRef]
  3. Mostofsky, E.; Penner, E.A.; Mittleman, M.A. Outbursts of anger as a trigger of acute cardiovascular events: A systematic review and meta-analysis. Eur. Heart J. 2014, 35, 1404–1410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Rozanski, A. Behavioral Cardiology: Current Advances and Future Directions. J. Am. Coll. Cardiol. 2014, 64, 100–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Vancheri, F.; Longo, G.; Vancheri, E.; Henein, M.Y. Mental Stress and Cardiovascular Health. J. Clin. Med. 2022, 11, 3353. [Google Scholar] [CrossRef] [PubMed]
  6. Brutsaert, D.L. Cardiac Endothelial-Myocardial Signaling: Its Role in Cardiac Growth, Contractile Performance, and Rhythmicity. Physiol. Rev. 2003, 83, 59–115. [Google Scholar] [CrossRef] [PubMed]
  7. Davignon, J.; Ganz, P. Role of Endothelial Dysfunction in Atherosclerosis. Circulation 2004, 109, III-27–III-32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Xue, Y.-T.; Tan, Q.-w.; Li, P.; Mou, S.-f.; Liu, S.-j.; Bao, Y.; Jiao, H.-c.; Su, W.-G. Investigating the role of acute mental stress on endothelial dysfunction: A systematic review and meta-analysis. Clin. Res. Cardiol. 2015, 104, 310–319. [Google Scholar] [CrossRef]
  9. Camici, P.G.; Crea, F. Coronary Microvascular Dysfunction. N. Engl. J. Med. 2007, 356, 830–840. [Google Scholar] [CrossRef] [Green Version]
  10. Gutiérrez, E.; Flammer, A.J.; Lerman, L.O.; Elízaga, J.; Lerman, A.; Fernández-Avilés, F. Endothelial dysfunction over the course of coronary artery disease. Eur. Heart J. 2013, 34, 3175–3181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Toda, N.; Nakanishi-Toda, M. How mental stress affects endothelial function. Pflügers Arch. Eur. J. Physiol. 2011, 462, 779–794. [Google Scholar] [CrossRef] [PubMed]
  12. Widlansky, M.E.; Gokce, N.; Keaney, J.F.; Vita, J.A. The clinical implications of endothelial dysfunction. J. Am. Coll. Cardiol. 2003, 42, 1149–1160. [Google Scholar] [CrossRef] [Green Version]
  13. Spicer, J.; Shimbo, D.; Johnston, N.; Harlapur, M.; Purdie-Vaughns, V.; Cook, J.; Fu, J.; Burg, M.M.; Wager, T.D. Prevention of Stress-Provoked Endothelial Injury by Values Affirmation: A Proof of Principle Study. Ann. Behav. Med. 2015, 50, 471–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Shimbo, D.; Rosenberg, L.B.; Chaplin, W.; Zhao, S.; Goldensohn, E.R.; Cholankeril, M.; Fu, J.; Hong, S.B.; Jelic, S.; Burg, M.M. Endothelial cell activation, reduced endothelial cell reparative capacity, and impaired endothelial-dependent vasodilation after anger provocation. Int. J. Cardiol. 2013, 167, 1064–1065. [Google Scholar] [CrossRef] [Green Version]
  15. Corretti, M.C.; Anderson, T.J.; Vallance, P.; Vita, J.; Vogel, R.; Benjamin, E.J.; Celermajer, D.; Charbonneau, F.; Creager, M.A.; Deanfield, J.; et al. Guidelines for the ultrasound assessment of endothelial-dependent flow-mediated vasodilation of the brachial artery: A report of the International Brachial Artery Reactivity Task Force. J. Am. Coll. Cardiol. 2002, 39, 257–265. [Google Scholar] [CrossRef] [Green Version]
  16. Poitras, V.J.; Pyke, K.E. The impact of acute mental stress on vascular endothelial function: Evidence, mechanisms and importance. Int. J. Psychophysiol. 2013, 88, 124–135. [Google Scholar] [CrossRef] [PubMed]
  17. Spieker, L.E.; Hürlimann, D.; Ruschitzka, F.; Corti, R.; Enseleit, F.; Shaw, S.; Hayoz, D.; Deanfield, J.E.; Lüscher, T.F.; Noll, G. Mental Stress Induces Prolonged Endothelial Dysfunction via Endothelin—A Receptors. Circulation 2002, 105, 2817–2820. [Google Scholar] [CrossRef] [Green Version]
  18. Broadley, A.J.M.; Korszun, A.; Abdelaal, E.; Moskvina, V.; Jones, C.J.H.; Nash, G.B.; Ray, C.; Deanfield, J.; Frenneaux, M.P. Inhibition of Cortisol Production with Metyrapone Prevents Mental Stress-Induced Endothelial Dysfunction and Baroreflex Impairment. J. Am. Coll. Cardiol. 2005, 46, 344–350. [Google Scholar] [CrossRef]
  19. Kivimäki, M.; Steptoe, A. Effects of stress on the development and progression of cardiovascular disease. Nat. Rev. Cardiol. 2018, 15, 215–229. [Google Scholar] [CrossRef] [PubMed]
  20. Ghiadoni, L.; Donald, A.E.; Cropley, M.; Mullen, M.J.; Oakley, G.; Taylor, M.; O’Connor, G.; Betteridge, J.; Klein, N.; Steptoe, A.; et al. Mental Stress Induces Transient Endothelial Dysfunction in Humans. Circulation 2000, 102, 2473–2478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Lima, B.B.; Hammadah, M.; Kim, J.H.; Uphoff, I.; Shah, A.; Levantsevych, O.; Almuwaqqat, Z.; Moazzami, K.; Sullivan, S.; Ward, L.; et al. Association of Transient Endothelial Dysfunction Induced by Mental Stress with Major Adverse Cardiovascular Events in Men and Women with Coronary Artery Disease. JAMA Cardiol. 2019, 4, 988–996. [Google Scholar] [CrossRef]
  22. Waclawovsky, A.J.; de Brito, E.; Smith, L.; Vancampfort, D.; da Silva, A.M.V.; Schuch, F.B. Endothelial dysfunction in people with depressive disorders: A systematic review and meta-analysis. J. Psychiatr. Res. 2021, 141, 152–159. [Google Scholar] [CrossRef] [PubMed]
  23. Hadi, H.A.R.; Carr, C.S.; Al Suwaidi, J. Endothelial dysfunction: Cardiovascular risk factors, therapy, and outcome. Vasc. Health Risk Manag. 2005, 1, 183–198. [Google Scholar] [PubMed]
  24. Bonetti, P.O.; Lerman, L.O.; Lerman, A. Endothelial dysfunction: A marker of atherosclerotic risk. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 168–175. [Google Scholar] [CrossRef]
  25. Aird, C.W. Endothelium as an organ system. Crit. Care Med. 2004, 32, S271–S279. [Google Scholar] [CrossRef]
  26. Lavi, S.; Bae, J.-H.; Rihal, C.S.; Prasad, A.; Barsness, G.W.; Lennon, R.J.; Holmes, D.R.; Lerman, A. Segmental coronary endothelial dysfunction in patients with minimal atherosclerosis is associated with necrotic core plaques. Heart 2009, 95, 1525–1530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Naghavi, M.; Libby, P.; Falk, E.; Casscells, S.W.; Litovsky, S.; Rumberger, J.; Badimon, J.J.; Stefanadis, C.; Moreno, P.; Pasterkamp, G.; et al. From Vulnerable Plaque to Vulnerable Patient: Part I. Circulation 2003, 108, 1664–1672. [Google Scholar] [CrossRef]
  28. Choi, B.-J.; Prasad, A.; Gulati, R.; Best, P.J.; Lennon, R.J.; Barsness, G.W.; Lerman, L.O.; Lerman, A. Coronary endothelial dysfunction in patients with early coronary artery disease is associated with the increase in intravascular lipid core plaque. Eur. Heart J. 2013, 34, 2047–2054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Gössl, M.; Yoon, M.-H.; Choi, B.-J.; Rihal, C.; Tilford, J.M.; Reriani, M.; Gulati, R.; Sandhu, G.; Eeckhout, E.; Lennon, R.; et al. Accelerated Coronary Plaque Progression and Endothelial Dysfunction: Serial Volumetric Evaluation by IVUS. JACC Cardiovasc. Imaging 2014, 7, 103–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Liu, M.; Liu, J.; Zhang, L.; Xu, W.; He, D.; Wei, W.; Ge, Y.; Dandu, C. An evidence of brain-heart disorder: Mental stress-induced myocardial ischemia regulated by inflammatory cytokines. Neurol. Res. 2020, 42, 670–675. [Google Scholar] [CrossRef] [PubMed]
  31. Paine, N.J.; Bosch, J.A.; Veldhuijzen Van Zanten, J.J.C.S. Inflammation and Vascular Responses to Acute Mental Stress: Implications for the Triggering of Myocardial Infarction. Curr. Pharm. Des. 2012, 18, 1494–1501. [Google Scholar] [CrossRef] [PubMed]
  32. Hammadah, M.; Sullivan, S.; Pearce, B.; Al Mheid, I.; Wilmot, K.; Ramadan, R.; Tahhan, A.S.; O’Neal, W.T.; Obideen, M.; Alkhoder, A.; et al. Inflammatory response to mental stress and mental stress induced myocardial ischemia. Brain Behav. Immun. 2018, 68, 90–97. [Google Scholar] [CrossRef] [PubMed]
  33. Kalogeropoulos, A.; Georgiopoulou, V.; Psaty, B.M.; Rodondi, N.; Smith, A.L.; Harrison, D.G.; Liu, Y.; Hoffmann, U.; Bauer, D.C.; Newman, A.B.; et al. Inflammatory Markers and Incident Heart Failure Risk in Older Adults: The Health ABC (Health, Aging, and Body Composition) Study. J. Am. Coll. Cardiol. 2010, 55, 2129–2137. [Google Scholar] [CrossRef] [Green Version]
  34. Kang, D.O.; Eo, J.S.; Park, E.J.; Nam, H.S.; Song, J.W.; Park, Y.H.; Park, S.Y.; Na, J.O.; Choi, C.U.; Kim, E.J.; et al. Stress-associated neurobiological activity is linked with acute plaque instability via enhanced macrophage activity: A prospective serial 18F-FDG-PET/CT imaging assessment. Eur. Heart J. 2021, 42, 1883–1895. [Google Scholar] [CrossRef]
  35. Moriya, J. Critical roles of inflammation in atherosclerosis. J. Cardiol. 2019, 73, 22–27. [Google Scholar] [CrossRef] [Green Version]
  36. Libby, P.; Tabas, I.; Fredman, G.; Fisher, E.A. Inflammation and its Resolution as Determinants of Acute Coronary Syndromes. Circ. Res. 2014, 114, 1867–1879. [Google Scholar] [CrossRef] [Green Version]
  37. Rohleder, N. Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom. Med. 2014, 76, 181–189. [Google Scholar] [CrossRef]
  38. Tawakol, A.; Ishai, A.; Takx, R.A.P.; Figueroa, A.L.; Ali, A.; Kaiser, Y.; Truong, Q.A.; Solomon, C.J.E.; Calcagno, C.; Mani, V.; et al. Relation between resting amygdalar activity and cardiovascular events: A longitudinal and cohort study. Lancet 2017, 389, 834–845. [Google Scholar] [CrossRef] [Green Version]
  39. Goyal, A.; Dey, A.K.; Chaturvedi, A.; Elnabawi, Y.A.; Aberra, T.M.; Chung, J.H.; Belur, A.D.; Groenendyk, J.W.; Lerman, J.B.; Rivers, J.P.; et al. Chronic Stress-Related Neural Activity Associates with Subclinical Cardiovascular Disease in Psoriasis: A Prospective Cohort Study. JACC Cardiovasc. Imaging 2020, 13, 465–477. [Google Scholar] [CrossRef]
  40. Gianaros, P.J.; Hariri, A.R.; Sheu, L.K.; Muldoon, M.F.; Sutton-Tyrrell, K.; Manuck, S.B. Preclinical Atherosclerosis Covaries with Individual Differences in Reactivity and Functional Connectivity of the Amygdala. Biol. Psychiatry 2009, 65, 943–950. [Google Scholar] [CrossRef] [Green Version]
  41. Muscatell, K.A.; Dedovic, K.; Slavich, G.M.; Jarcho, M.R.; Breen, E.C.; Bower, J.E.; Irwin, M.R.; Eisenberger, N.I. Greater amygdala activity and dorsomedial prefrontal–amygdala coupling are associated with enhanced inflammatory responses to stress. Brain Behav. Immun. 2015, 43, 46–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Tracey, K.J. The inflammatory reflex. Nature 2002, 420, 853–859. [Google Scholar] [CrossRef] [PubMed]
  43. Borovikova, L.V.; Ivanova, S.; Zhang, M.; Yang, H.; Botchkina, G.I.; Watkins, L.R.; Wang, H.; Abumrad, N.; Eaton, J.W.; Tracey, K.J. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000, 405, 458–462. [Google Scholar] [CrossRef] [PubMed]
  44. Kop, W.J.; Weissman, N.J.; Zhu, J.; Bonsall, R.W.; Doyle, M.; Stretch, M.R.; Glaes, S.B.; Krantz, D.S.; Gottdiener, J.S.; Tracy, R.P. Effects of Acute Mental Stress and Exercise on Inflammatory Markers in Patients with Coronary Artery Disease and Healthy Controls. Am. J. Cardiol. 2008, 101, 767–773. [Google Scholar] [CrossRef] [PubMed]
  45. Pavlov, V.A.; Tracey, K.J. The vagus nerve and the inflammatory reflex—Linking immunity and metabolism. Nat. Rev. Endocrinol. 2012, 8, 743–754. [Google Scholar] [CrossRef]
  46. Olshansky, B. Vagus nerve modulation of inflammation: Cardiovascular implications. Trends Cardiovasc. Med. 2016, 26, 1–11. [Google Scholar] [CrossRef]
  47. Wirtz, P.H.; von Känel, R. Psychological Stress, Inflammation, and Coronary Heart Disease. Curr. Cardiol. Rep. 2017, 19, 1–10. [Google Scholar] [CrossRef]
  48. Steptoe, A.; Hamer, M.; Chida, Y. The effects of acute psychological stress on circulating inflammatory factors in humans: A review and meta-analysis. Brain Behav. Immun. 2007, 21, 901–912. [Google Scholar] [CrossRef]
  49. Burg, M.M.; Soufer, A.; Lampert, R.; Collins, D.; Soufer, R. Autonomic Contribution to Endothelin-1 Increase during Laboratory Anger-Recall Stress in Patients with Coronary Artery Disease. Mol. Med. 2011, 17, 495–501. [Google Scholar] [CrossRef]
  50. The Emerging Risk Factors Collaboration. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: A collaborative meta-analysis of 102 prospective studies. Lancet 2010, 375, 2215–2222. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, W.; Speiser, J.L.; Ye, F.; Tsai, M.Y.; Cainzos-Achirica, M.; Nasir, K.; Herrington, D.M.; Shapiro, M.D. High-Sensitivity C-Reactive Protein Modifies the Cardiovascular Risk of Lipoprotein(a): Multi-Ethnic Study of Atherosclerosis. J. Am. Coll. Cardiol. 2021, 78, 1083–1094. [Google Scholar] [CrossRef] [PubMed]
  52. Koenig, W. C-Reactive Protein and Cardiovascular Risk: Will the Controversy End after CANTOS? Clin. Chem. 2017, 63, 1897–1898. [Google Scholar] [CrossRef] [PubMed]
  53. Koenig, W. High-sensitivity C-reactive protein and atherosclerotic disease: From improved risk prediction to risk-guided therapy. Int. J. Cardiol. 2013, 168, 5126–5134. [Google Scholar] [CrossRef]
  54. Heidt, T.; Sager, H.B.; Courties, G.; Dutta, P.; Iwamoto, Y.; Zaltsman, A.; von Zur Muhlen, C.; Bode, C.; Fricchione, G.L.; Denninger, J.; et al. Chronic variable stress activates hematopoietic stem cells. Nat. Med. 2014, 20, 754–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hinterdobler, J.; Schott, S.; Jin, H.; Meesmann, A.; Steinsiek, A.-L.; Zimmermann, A.-S.; Wobst, J.; Müller, P.; Mauersberger, C.; Vilne, B.; et al. Acute mental stress drives vascular inflammation and promotes plaque destabilization in mouse atherosclerosis. Eur. Heart J. 2021, 42, 4077–4088. [Google Scholar] [CrossRef] [PubMed]
  56. Hanna, R.N.; Hedrick, C.C. Stressing out stem cells: Linking stress and hematopoiesis in cardiovascular disease. Nat. Med. 2014, 20, 707–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. van der Heijden Charlotte, D.C.C.; Groh, L.; Keating, S.T.; Kaffa, C.; Noz, M.P.; Kersten, S.; Herwaarden, A.E.v.; Hoischen, A.; Joosten, L.A.B.; Timmers, H.J.L.M.; et al. Catecholamines Induce Trained Immunity in Monocytes In Vitro and In Vivo. Circ. Res. 2020, 127, 269–283. [Google Scholar] [CrossRef]
  58. Swirski, F.K.; Nahrendorf, M. Leukocyte Behavior in Atherosclerosis, Myocardial Infarction, and Heart Failure. Science 2013, 339, 161–166. [Google Scholar] [CrossRef] [Green Version]
  59. Moore, K.J.; Tabas, I. Macrophages in the Pathogenesis of Atherosclerosis. Cell 2011, 145, 341–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Libby, P. Mechanisms of Acute Coronary Syndromes and Their Implications for Therapy. N. Engl. J. Med. 2013, 368, 2004–2013. [Google Scholar] [CrossRef] [Green Version]
  61. Lagraauw, H.M.; Wezel, A.; van der Velden, D.; Kuiper, J.; Bot, I. Stress-induced mast cell activation contributes to atherosclerotic plaque destabilization. Sci. Rep. 2019, 9, 2134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Eryd, S.A.; Smith, J.G.; Melander, O.; Hedblad, B.; Engström, G. Incidence of Coronary Events and Case Fatality Rate in Relation to Blood Lymphocyte and Neutrophil Counts. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 533–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Marsland, A.L.; Walsh, C.; Lockwood, K.; John-Henderson, N.A. The effects of acute psychological stress on circulating and stimulated inflammatory markers: A systematic review and meta-analysis. Brain Behav. Immun. 2017, 64, 208–219. [Google Scholar] [CrossRef] [PubMed]
  64. Lagraauw, H.M.; Kuiper, J.; Bot, I. Acute and chronic psychological stress as risk factors for cardiovascular disease: Insights gained from epidemiological, clinical and experimental studies. Brain Behav. Immun. 2015, 50, 18–30. [Google Scholar] [CrossRef]
  65. Walker, B.R. Glucocorticoids and Cardiovascular Disease. Eur. J. Endocrinol. 2007, 157, 545–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hamer, M.; Endrighi, R.; Venuraju, S.M.; Lahiri, A.; Steptoe, A. Cortisol Responses to Mental Stress and the Progression of Coronary Artery Calcification in Healthy Men and Women. PLoS ONE 2012, 7, e31356. [Google Scholar] [CrossRef] [PubMed]
  67. Black, P.H.; Garbutt, L.D. Stress, inflammation and cardiovascular disease. J. Psychosom. Res. 2002, 52, 1–23. [Google Scholar] [CrossRef]
  68. Nijm, J.; Kristenson, M.; Olsson, A.G.; Jonasson, L. Impaired cortisol response to acute stressors in patients with coronary disease. Implications for inflammatory activity. J. Intern. Med. 2007, 262, 375–384. [Google Scholar] [CrossRef]
  69. Brydon, L.; Magid, K.; Steptoe, A. Platelets, coronary heart disease, and stress. Brain Behav. Immun. 2006, 20, 113–119. [Google Scholar] [CrossRef]
  70. Bytyçi, I.; Bajraktari, G.; Penson, P.E.; Henein, M.Y.; Banach, M. Efficacy and safety of colchicine in patients with coronary artery disease: A systematic review and meta-analysis of randomized controlled trials. Br. J. Clin. Pharmacol. 2021, 88, 1520–1528. [Google Scholar] [CrossRef] [PubMed]
  71. Andreis, A.; Imazio, M.; Piroli, F.; Avondo, S.; Casula, M.; Paneva, E.; De Ferrari, G.M. Efficacy and safety of colchicine for the prevention of major cardiovascular and cerebrovascular events in patients with coronary artery disease: A systematic review and meta-analysis on 12,869 patients. Eur. J. Prev. Cardiol. 2021, 28, 1916–1925. [Google Scholar] [CrossRef] [PubMed]
  72. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D. Antiinflammatory therapy with canakinumab for atherosclerotic disease. New Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  73. Steptoe, A.; Kivimäki, M. Stress and Cardiovascular Disease: An Update on Current Knowledge. Annu. Rev. Public Health 2013, 34, 337–354. [Google Scholar] [CrossRef] [PubMed]
  74. Esler, M. Mental stress and human cardiovascular disease. Neurosci. Biobehav. Rev. 2017, 74, 269–276. [Google Scholar] [CrossRef] [PubMed]
  75. Kivimäki, M.; Jokela, M.; Nyberg, S.T.; Singh-Manoux, A.; Fransson, E.I.; Alfredsson, L.; Bjorner, J.B.; Borritz, M.; Burr, H.; Casini, A.; et al. Long working hours and risk of coronary heart disease and stroke: A systematic review and meta-analysis of published and unpublished data for 603,838 individuals. Lancet 2015, 386, 1739–1746. [Google Scholar] [CrossRef] [Green Version]
  76. Bergmann, N.; Gyntelberg, F.; Faber, J. The appraisal of chronic stress and the development of the metabolic syndrome: A systematic review of prospective cohort studies. Endocr. Connect. 2014, 3, R55–R80. [Google Scholar] [CrossRef] [PubMed]
  77. Spruill, T.M. Chronic Psychosocial Stress and Hypertension. Curr. Hypertens. Rep. 2010, 12, 10–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Liu, M.-Y.; Li, N.; Li, W.A.; Khan, H. Association between psychosocial stress and hypertension: A systematic review and meta-analysis. Neurol. Res. 2017, 39, 573–580. [Google Scholar] [CrossRef] [PubMed]
  79. Esler, M.; Eikelis, N.; Schlaich, M.; Lambert, G.; Alvarenga, M.; Dawood, T.; Kaye, D.; Barton, D.; Pier, C.; Guo, L.; et al. Chronic mental stress is a cause of essential hypertension: Presence of biological markers of stress. Clin. Exp. Pharmacol. Physiol. 2008, 35, 498–502. [Google Scholar] [CrossRef] [PubMed]
  80. Oparil, S.; Zaman, M.A.; Calhoun, D.A. Pathogenesis of hypertension. Ann. Intern. Med. 2003, 139, 761–776. [Google Scholar] [CrossRef] [PubMed]
  81. Hamer, M.; Steptoe, A. Cortisol Responses to Mental Stress and Incident Hypertension in Healthy Men and Women. J. Clin. Endocrinol. Metab. 2012, 97, E29–E34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Carroll, D.; Ginty, A.T.; Der, G.; Hunt, K.; Benzeval, M.; Phillips, A.C. Increased blood pressure reactions to acute mental stress are associated with 16-year cardiovascular disease mortality. Psychophysiology 2012, 49, 1444–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Matthews, K.A.; Katholi, C.R.; McCreath, H.; Whooley, M.A.; Williams, D.R.; Zhu, S.; Markovitz, J.H. Blood pressure reactivity to psychological stress predicts hypertension in the CARDIA study. Circulation 2004, 110, 74–78. [Google Scholar] [CrossRef] [PubMed]
  84. Steptoe, A.; Kivimäki, M.; Lowe, G.; Rumley, A.; Hamer, M. Blood Pressure and Fibrinogen Responses to Mental Stress as Predictors of Incident Hypertension over an 8-Year Period. Ann. Behav. Med. 2016, 50, 898–906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Jennings, J.R.; Kamarck, T.W.; Everson-Rose, S.A.; Kaplan, G.A.; Manuck, S.B.; Salonen, J.T. Exaggerated blood pressure responses during mental stress are prospectively related to enhanced carotid atherosclerosis in middle-aged Finnish men. Circulation 2004, 110, 2198–2203. [Google Scholar] [CrossRef] [Green Version]
  86. Chida, Y.; Steptoe, A. Greater Cardiovascular Responses to Laboratory Mental Stress Are Associated with Poor Subsequent Cardiovascular Risk Status: A Meta-Analysis of Prospective Evidence. Hypertension 2010, 55, 1026–1032. [Google Scholar] [CrossRef] [Green Version]
  87. Barnett, P.A.; Spence, J.D.; Manuck, S.B.; Jennings, J.R. Psychological stress and the progression of carotid artery disease. J. Hypertens. 1997, 15, 49–55. [Google Scholar] [CrossRef] [PubMed]
  88. Paterniti, S.; Zureik, M.; Ducimetière, P.; Touboul, P.-J.; Fève, J.-M.; Alpérovitch, A. Sustained Anxiety and 4-Year Progression of Carotid Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 136–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Hemingway, H.; Malik, M.; Marmot, M. Social and psychosocial influences on sudden cardiac death, ventricular arrhythmia and cardiac autonomic function. Eur. Heart J. 2001, 22, 1082–1101. [Google Scholar] [CrossRef] [Green Version]
  90. Batelaan, N.M.; Seldenrijk, A.; van den Heuvel, O.A.; van Balkom, A.J.L.M.; Kaiser, A.; Reneman, L.; Tan, H.L. Anxiety, Mental Stress, and Sudden Cardiac Arrest: Epidemiology, Possible Mechanisms and Future Research. Front. Psychiatry 2022, 12, 813518. [Google Scholar] [CrossRef]
  91. Lampert, R. Mental Stress and Ventricular Arrhythmias. Curr. Cardiol. Rep. 2016, 18, 118. [Google Scholar] [CrossRef] [PubMed]
  92. Lampert, R.; Joska, T.; Burg, M.M.; Batsford, W.P.; McPherson, C.A.; Jain, D. Emotional and physical precipitants of ventricular arrhythmia. Circulation 2002, 106, 1800–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Rosman, L.; Lampert, R.; Ramsey, C.M.; Dziura, J.; Chui, P.W.; Brandt, C.; Haskell, S.; Burg, M.M. Posttraumatic Stress Disorder and Risk for Early Incident Atrial Fibrillation: A Prospective Cohort Study of 1.1 Million Young Adults. J. Am. Heart Assoc. 2019, 8, e013741. [Google Scholar] [CrossRef] [Green Version]
  94. Cheng, Y.-F.; Leu, H.-B.; Su, C.-C.; Huang, C.-C.; Chiang, C.-H.; Huang, P.-H.; Chung, C.-M.; Lin, S.-J.; Chen, J.-W.; Chan, W.-L. Association between panic disorder and risk of atrial fibrillation:a nationwide study. Psychosom. Med. 2013, 75, 30–35. [Google Scholar] [CrossRef] [PubMed]
  95. Shen, Z.-X.; Sun, Y.-M.; Gu, H.-H.; Zhang, Y.; Shen, Z.-W.; Liang, X.-N.; Ding, D.; Wang, J. Association between anxiety symptoms and atrial fibrillation in a community cohort of Chinese older adults: A case-control study. BMC Cardiovasc. Disord. 2021, 21, 471. [Google Scholar] [CrossRef] [PubMed]
  96. Winbo, A.; Paterson, D.J. The Brain-Heart Connection in Sympathetically Triggered Inherited Arrhythmia Syndromes. Heart Lung Circ. 2020, 29, 529–537. [Google Scholar] [CrossRef] [Green Version]
  97. Lankaputhra, M.; Voskoboinik, A. Congenital long QT syndrome: A clinician’s guide. Intern. Med. J. 2021, 51, 1999–2011. [Google Scholar] [CrossRef]
  98. Ylänen, K.; Poutanen, T.; Hiippala, A.; Swan, H.; Korppi, M. Catecholaminergic polymorphic ventricular tachycardia. Eur. J. Pediatrics 2010, 169, 535–542. [Google Scholar] [CrossRef]
  99. Paavonen, K.J.; Swan, H.; Piippo, K.; Hokkanen, L.; Laitinen, P.; Viitasalo, M.; Toivonen, L.; Kontula, K. Response of the QT interval to mental and physical stress in types LQT1 and LQT2 of the long QT syndrome. Heart 2001, 86, 39–44. [Google Scholar] [CrossRef]
  100. Etienne, P.; Huchet, F.; Gaborit, N.; Barc, J.; Thollet, A.; Kyndt, F.; Guyomarch, B.; Le Marec, H.; Charpentier, F.; Schott, J.-J.; et al. Mental stress test: A rapid, simple, and efficient test to unmask long QT syndrome. Europace 2018, 20, 2014–2020. [Google Scholar] [CrossRef]
  101. Skinner, J.R.; Winbo, A.; Abrams, D.; Vohra, J.; Wilde, A.A. Channelopathies That Lead to Sudden Cardiac Death: Clinical and Genetic Aspects. Heart Lung Circ. 2019, 28, 22–30. [Google Scholar] [CrossRef] [PubMed]
  102. Finlay, M.C.M.P.; Lambiase, P.D.P.F.F.; Ben-Simon, R.M.D.F.; Taggart, P.D.F. Effect of mental stress on dynamic electrophysiological properties of the endocardium and epicardium in humans. Heart Rhythm. 2016, 13, 175–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Herring, N.; Kalla, M.; Paterson, D.J. The autonomic nervous system and cardiac arrhythmias: Current concepts and emerging therapies. Nat. Rev. Cardiol. 2019, 16, 707–726. [Google Scholar] [CrossRef] [PubMed]
  104. Qu, Z.; Xie, L.-H.; Olcese, R.; Karagueuzian, H.S.; Chen, P.-S.; Garfinkel, A.; Weiss, J.N. Early afterdepolarizations in cardiac myocytes: Beyond reduced repolarization reserve. Cardiovasc. Res. 2013, 99, 6–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Huang, X.; Song, Z.; Qu, Z. Determinants of early afterdepolarization properties in ventricular myocyte models. PLoS Comput. Biol. 2018, 14, e1006382. [Google Scholar] [CrossRef] [Green Version]
  106. Leite, L.R.; Henz, B.D.; Macedo, P.G.; Santos, S.N.; Barreto, J.R.; Zanatta, A.; Fenelon, G.; Cruz Filho, F.E.S. Catecholaminergic polymorphic ventricular tachycardia: A current overview. Future Cardiol. 2009, 5, 191–199. [Google Scholar] [CrossRef]
  107. Tse, G.M.A.M.P. Mechanisms of cardiac arrhythmias. J. Arrhythmia 2015, 32, 75–81. [Google Scholar] [CrossRef] [Green Version]
  108. Ackerman, M.J.M.D.P.; Priori, S.G.M.D.P.; Dubin, A.M.D.F.; Kowey, P.M.D.; Linker, N.J.M.D.F.; Slotwiner, D.M.D.F.; Triedman, J.M.D.F.C.C.; Van Hare, G.F.M.D.F.C.C.; Gold, M.R.M.D.P.F. Beta Blocker Therapy for Long QT Syndrome and Catecholaminergic Polymorphic Ventricular Tachycardia: Are all beta blockers equivalent? Heart Rhythm. 2016, 14, e41–e44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Priori, S.G.; Blomström-Lundqvist, C.; Mazzanti, A.; Blom, N.; Borggrefe, M.; Camm, J.; Elliott, P.M.; Fitzsimons, D.; Hatala, R.; Hindricks, G.; et al. 2015 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: The Task Force for the Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death of the European Society of Cardiology (ESC). Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC). Eur. Heart J. 2015, 36, 2793–2867. [Google Scholar] [CrossRef] [Green Version]
  110. Sanghai, S.; Abbott, N.J.; Dewland, T.A.; Henrikson, C.A.; Elman, M.R.; Wollenberg, M.; Ivie, R.; Gonzalez-Sotomayor, J.; Nazer, B. Stellate Ganglion Blockade with Continuous Infusion Versus Single Injection for Treatment of Ventricular Arrhythmia Storm. Clin. Electrophysiol. 2021, 7, 452–460. [Google Scholar] [CrossRef] [PubMed]
  111. Lampert, R.; Shusterman, V.; Burg, M.; McPherson, C.; Batsford, W.; Goldberg, A.; Soufer, R. Anger-Induced T-Wave Alternans Predicts Future Ventricular Arrhythmias in Patients with Implantable Cardioverter-Defibrillators. J. Am. Coll. Cardiol. 2009, 53, 774–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lane, R.D.; Jennings, J.R. Hemispheric asymmetry, autonomic asymmetry and the problem of sudden cardiac death. In Brain asymmetry; Davidson, R.J.H.K., Ed.; MIT Press: Cambridge, MA, USA, 1995; pp. 271–304. [Google Scholar]
  113. Critchley, H.D.; Taggart, P.; Sutton, P.M.; Holdright, D.R.; Batchvarov, V.; Hnatkova, K.; Malik, M.; Dolan, R.J. Mental stress and sudden cardiac death: Asymmetric midbrain activity as a linking mechanism. Brain 2005, 128, 75–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ruiz Vargas, E.; Sörös, P.; Shoemaker, J.K.; Hachinski, V. Human cerebral circuitry related to cardiac control: A neuroimaging meta-analysis. Ann. Neurol. 2016, 79, 709–716. [Google Scholar] [CrossRef] [PubMed]
  115. Chouchou, F.; Mauguière, F.; Vallayer, O.; Catenoix, H.; Isnard, J.; Montavont, A.; Jung, J.; Pichot, V.; Rheims, S.; Mazzola, L. How the insula speaks to the heart: Cardiac responses to insular stimulation in humans. Hum. Brain Mapp. 2019, 40, 2611–2622. [Google Scholar] [CrossRef] [PubMed]
  116. Taggart, P.; Critchley, H.; Lambiase, P.D. Heart–brain interactions in cardiac arrhythmia. Heart 2011, 97, 698–708. [Google Scholar] [CrossRef] [PubMed]
  117. Peacock, J.; Whang, W. Psychological Distress and Arrhythmia: Risk Prediction and Potential Modifiers. Prog. Cardiovasc. Dis. 2013, 55, 582–589. [Google Scholar] [CrossRef] [Green Version]
  118. Ghadri, J.-R.; Wittstein, I.S.; Prasad, A.; Sharkey, S.; Dote, K.; Akashi, Y.J.; Cammann, V.L.; Crea, F.; Galiuto, L.; Desmet, W.; et al. International Expert Consensus Document on Takotsubo Syndrome (Part I): Clinical Characteristics, Diagnostic Criteria, and Pathophysiology. Eur. Heart J. 2018, 39, 2032–2046. [Google Scholar] [CrossRef] [Green Version]
  119. Ako, J.; Sudhir, K.; Farouque, H.M.O.; Honda, Y.; Fitzgerald, P.J. Transient Left Ventricular Dysfunction under Severe Stress: Brain-Heart Relationship Revisited. Am. J. Med. 2006, 119, 10–17. [Google Scholar] [CrossRef] [PubMed]
  120. Osawa, A.; Nagai, M.; Dote, K.; Kato, M.; Oda, N.; Kunita, E.; Kagawa, E.; Yamane, A.; Kobatake, H.; Shiota, H.; et al. A mid-ventricular variant of Takotsubo syndrome: Was it triggered by insular cortex damage? ESC Heart Fail. 2021, 8, 3408–3412. [Google Scholar] [CrossRef]
  121. Akashi, Y.J.; Nef, H.M.; Lyon, A.R. Epidemiology and pathophysiology of Takotsubo syndrome. Nat. Rev. Cardiol. 2015, 12, 387–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Templin, C.; Ghadri, J.R.; Diekmann, J.; Napp, L.C.; Bataiosu, D.R.; Jaguszewski, M.; Cammann, V.L.; Sarcon, A.; Geyer, V.; Neumann, C.A.; et al. Clinical Features and Outcomes of Takotsubo (Stress) Cardiomyopathy. N. Engl. J. Med. 2015, 373, 929–938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Sharkey, S.W.; Lesser, J.R.; Maron, M.S.; Maron, B.J. Why Not Just Call It Tako-Tsubo Cardiomyopathy: A Discussion of Nomenclature. J. Am. Coll. Cardiol. 2011, 57, 1496–1497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Gopinath, R.; Ayya, S.S. Neurogenic stress cardiomyopathy: What do we need to know. Ann. Card. Anaesth. 2018, 21, 228–234. [Google Scholar] [CrossRef] [PubMed]
  125. Summers, M.R.; Lennon, R.J.; Prasad, A. Pre-Morbid Psychiatric and Cardiovascular Diseases in Apical Ballooning Syndrome (Tako-Tsubo/Stress-Induced Cardiomyopathy): Potential Pre-Disposing Factors? J. Am. Coll. Cardiol. 2010, 55, 700–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Ghadri, J.R.; Sarcon, A.; Diekmann, J.; Bataiosu, D.R.; Cammann, V.L.; Jurisic, S.; Napp, L.C.; Jaguszewski, M.; Scherff, F.; Brugger, P.; et al. Happy heart syndrome: Role of positive emotional stress in takotsubo syndrome. Eur. Heart J. 2016, 37, 2823–2829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Nagai, M.; Kobayashi, Y.; Kobatake, H.; Dote, K.; Kato, M.; Oda, N.; Kunita, E.; Kagawa, E.; Yamane, A.; Osawa, A.; et al. Happy heart syndrome: A case of Takotsubo syndrome with left internal carotid artery occlusion. Clin. Auton. Res. 2020, 30, 347–350. [Google Scholar] [CrossRef] [PubMed]
  128. Esslen, M.; Pascual-Marqui, R.D.; Hell, D.; Kochi, K.; Lehmann, D. Brain areas and time course of emotional processing. NeuroImage 2004, 21, 1189–1203. [Google Scholar] [CrossRef] [PubMed]
  129. Kaski, J.C. Cardiac syndrome X in women: The role of oestrogen deficiency. Heart 2006, 92, iii5–iii9. [Google Scholar] [CrossRef] [PubMed]
  130. Sader, M.A.; Celermajer, D.S. Endothelial function, vascular reactivity and gender differences in the cardiovascular system. Cardiovasc. Res. 2002, 53, 597–604. [Google Scholar] [CrossRef]
  131. Sung, B.H.; Ching, M.; Izzo, J.L.; Dandona, P.; Wilson, M.F. Estrogen improves abnormal norepinephrine-induced vasoconstriction in postmenopausal women. J. Hypertens. 1999, 17, 523–528. [Google Scholar] [CrossRef] [PubMed]
  132. Vitale, C.; Mendelsohn, M.E.; Rosano, G.M.C. Gender differences in the cardiovascular effect of sex hormones. Nat. Rev. Cardiol. 2009, 6, 532–542. [Google Scholar] [CrossRef] [PubMed]
  133. Pelliccia, F.; Kaski, J.C.; Crea, F.; Camici, P.G. Pathophysiology of Takotsubo Syndrome. Circulation 2017, 135, 2426–2441. [Google Scholar] [CrossRef]
  134. Scally, C.; Abbas, H.; Ahearn, T.; Srinivasan, J.; Mezincescu, A.; Rudd, A.; Spath, N.; Yucel-Finn, A.; Yuecel, R.; Oldroyd, K.; et al. Myocardial and Systemic Inflammation in Acute Stress-Induced (Takotsubo) Cardiomyopathy. Circulation 2019, 139, 1581–1592. [Google Scholar] [CrossRef] [PubMed]
  135. Lüscher, T.F.; Templin, C. Is takotsubo syndrome a microvascular acute coronary syndrome? Towards of a new definition. Eur. Heart J. 2016, 37, 2816–2820. [Google Scholar] [CrossRef] [PubMed]
  136. Moloney, R.D.; Johnson, A.C.; O’Mahony, S.M.; Dinan, T.G.; Greenwood-Van Meerveld, B.; Cryan, J.F. Stress and the Microbiota–Gut–Brain Axis in Visceral Pain: Relevance to Irritable Bowel Syndrome. CNS Neurosci. Ther. 2016, 22, 102–117. [Google Scholar] [CrossRef]
  137. Templin, C.; Hänggi, J.; Klein, C.; Topka, M.S.; Hiestand, T.; Levinson, R.A.; Jurisic, S.; Lüscher, T.F.; Ghadri, J.-R.; Jäncke, L. Altered limbic and autonomic processing supports brain-heart axis in Takotsubo syndrome. Eur. Heart J. 2019, 40, 1183–1187. [Google Scholar] [CrossRef]
  138. Hiestand, T.; Hänggi, J.; Klein, C.; Topka, M.S.; Jaguszewski, M.; Ghadri, J.R.; Lüscher, T.F.; Jäncke, L.; Templin, C. Takotsubo Syndrome Associated with Structural Brain Alterations of the Limbic System. J. Am. Coll. Cardiol. 2018, 71, 809–811. [Google Scholar] [CrossRef]
  139. Silva, A.R.; Magalhães, R.; Arantes, C.; Moreira, P.S.; Rodrigues, M.; Marques, P.; Marques, J.; Sousa, N.; Pereira, V.H. Brain functional connectivity is altered in patients with Takotsubo Syndrome. Sci. Rep. 2019, 9, 4187. [Google Scholar] [CrossRef] [Green Version]
  140. Dichtl, W.; Tuovinen, N.; Barbieri, F.; Adukauskaite, A.; Senoner, T.; Rubatscher, A.; Hintringer, F.; Siedentopf, C.; Bauer, A.; Gizewski, E.R.; et al. Functional neuroimaging in the acute phase of Takotsubo syndrome: Volumetric and functional changes of the right insular cortex. Clin. Res. Cardiol. 2020, 109, 1107–1113. [Google Scholar] [CrossRef] [Green Version]
  141. Klein, C.; Hiestand, T.; Ghadri, J.-R.; Templin, C.; Jäncke, L.; Hänggi, J. Takotsubo Syndrome—Predictable from brain imaging data. Sci. Rep. 2017, 7, 5434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Sciagrà, R.; Parodi, G.; Del Pace, S.; Genovese, S.; Zampini, L.; Bellandi, B.; Gensini, G.F.; Pupi, A.; Antoniucci, D. Abnormal response to mental stress in patients with Takotsubo cardiomyopathy detected by gated single photon emission computed tomography. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 765–772. [Google Scholar] [CrossRef] [PubMed]
  143. Radfar, A.; Abohashem, S.; Osborne, M.T.; Wang, Y.; Dar, T.; Hassan, M.Z.O.; Ghoneem, A.; Naddaf, N.; Patrich, T.; Abbasi, T.; et al. Stress-associated neurobiological activity associates with the risk for and timing of subsequent Takotsubo syndrome. Eur. Heart J. 2021, 42, 1898–1907. [Google Scholar] [CrossRef] [PubMed]
  144. Samad, Z.; Boyle, S.; Ersboll, M.; Vora, A.N.; Zhang, Y.; Becker, R.C.; Williams, R.; Kuhn, C.; Ortel, T.L.; Rogers, J.G.; et al. Sex Differences in Platelet Reactivity and Cardiovascular and Psychological Response to Mental Stress in Patients with Stable Ischemic Heart Disease: Insights from the REMIT Study. J. Am. Coll. Cardiol. 2014, 64, 1669–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Vaccarino, V.; Wilmot, K.; Mheid, I.A.; Ramadan, R.; Pimple, P.; Shah, A.J.; Garcia, E.V.; Nye, J.; Ward, L.; Hammadah, M.; et al. Sex Differences in Mental Stress-Induced Myocardial Ischemia in Patients with Coronary Heart Disease. J. Am. Heart Assoc. 2016, 5, e003630. [Google Scholar] [CrossRef]
  146. Wokhlu, A.; Pepine, C.J. Mental Stress and Myocardial Ischemia: Young Women at Risk. J. Am. Heart Assoc. 2016, 5, e004196. [Google Scholar] [CrossRef]
  147. Soufer, R.; Jain, H.; Yoon, A.J. Heart-brain interactions in mental stress-induced myocardial ischemia. Curr. Cardiol. Rep. 2009, 11, 133–140. [Google Scholar] [CrossRef] [Green Version]
  148. Kasher, N.; Wittbrodt, M.T.; Alam, Z.S.; Lima, B.B.; Nye, J.A.; Campanella, C.; Ladd, S.; Hammadah, M.; Shah, A.J.; Raggi, P.; et al. Sex differences in brain activation patterns with mental stress in patients with coronary artery disease. Biol. Sex Differ. 2019, 10, 35. [Google Scholar] [CrossRef] [Green Version]
  149. Sullivan, S.; Hammadah, M.; Al Mheid, I.; Wilmot, K.; Ramadan, R.; Alkhoder, A.; Isakadze, N.; Shah, A.; Levantsevych, O.; Pimple, P.M.; et al. Sex Differences in Hemodynamic and Microvascular Mechanisms of Myocardial Ischemia Induced by Mental Stress. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 473–480. [Google Scholar] [CrossRef] [Green Version]
  150. Martin, E.A.; Tan, S.-L.; MacBride, L.R.; Lavi, S.; Lerman, L.O.; Lerman, A. Sex differences in vascular and endothelial responses to acute mental stress. Clin. Auton. Res. 2008, 18, 339–345. [Google Scholar] [CrossRef] [Green Version]
  151. Reis, S.E.; Holubkov, R.; Smith, A.J.C.; Kelsey, S.F.; Sharaf, B.L.; Reichek, N.; Rogers, W.J.; Merz, C.N.B.; Sopko, G.; Pepine, C.J. Coronary microvascular dysfunction is highly prevalent in women with chest pain in the absence of coronary artery disease: Results from the NHLBI WISE study. Am. Heart J. 2001, 141, 735–741. [Google Scholar] [CrossRef] [Green Version]
  152. Masi, C.M.; Chen, H.-Y.; Hawkley, L.C.; Cacioppo, J.T. A Meta-Analysis of Interventions to Reduce Loneliness. Personal. Soc. Psychol. Rev. 2011, 15, 219–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Gulliksson, M.; Burell, G.; Vessby, B.; Lundin, L.; Toss, H.; Svärdsudd, K. Randomized Controlled Trial of Cognitive Behavioral Therapy vs. Standard Treatment to Prevent Recurrent Cardiovascular Events in Patients with Coronary Heart Disease: Secondary Prevention in Uppsala Primary Health Care Project (SUPRIM). Arch. Intern. Med. 2011, 171, 134–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Orth-Gomér, K.; Schneiderman, N.; Wang, H.-X.; Walldin, C.; Blom, M.; Jernberg, T. Stress reduction prolongs life in women with coronary disease: The Stockholm Women’s Intervention Trial for Coronary Heart Disease (SWITCHD). Circ. Cardiovasc. Qual. Outcomes 2009, 2, 25–32. [Google Scholar] [CrossRef] [Green Version]
  155. Kim, J.-M.; Stewart, R.; Lee, Y.-S.; Lee, H.-J.; Kim, M.C.; Kim, J.-W.; Kang, H.-J.; Bae, K.-Y.; Kim, S.-W.; Shin, I.-S.; et al. Effect of Escitalopram vs. Placebo Treatment for Depression on Long-Term Cardiac Outcomes in Patients with Acute Coronary Syndrome: A Randomized Clinical Trial. JAMA 2018, 320, 350–357. [Google Scholar] [CrossRef] [Green Version]
  156. Stewart, J.C.; Perkins, A.J.; Callahan, C.M. Effect of collaborative care for depression on risk of cardiovascular events: Data from the IMPACT randomized controlled trial. Psychosom. Med. 2014, 76, 29–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Jiang, W.; Velazquez, E.J.; Kuchibhatla, M.; Samad, Z.; Boyle, S.H.; Kuhn, C.; Becker, R.C.; Ortel, T.L.; Williams, R.B.; Rogers, J.G.; et al. Effect of Escitalopram on Mental Stress–Induced Myocardial Ischemia: Results of the REMIT Trial. JAMA 2013, 309, 2139–2149. [Google Scholar] [CrossRef] [PubMed]
  158. Glassman, A.H.; O’Connor, C.M.; Califf, R.M.; Swedberg, K.; Schwartz, P.; Bigger, J.J.T.; Krishnan, K.R.R.; van Zyl, L.T.; Swenson, J.R.; Finkel, M.S.; et al. Sertraline Treatment of Major Depression in Patients with Acute MI or Unstable Angina. JAMA 2002, 288, 701–709. [Google Scholar] [CrossRef]
  159. Berkman, L.F.; Blumenthal, J.; Burg, M.; Carney, R.M.; Catellier, D.; Cowan, M.J.; Czajkowski, S.M.; DeBusk, R.; Hosking, J.; Jaffe, A.; et al. Effects of Treating Depression and Low Perceived Social Support on Clinical Events after Myocardial Infarction: The Enhancing Recovery in Coronary Heart Disease Patients (ENRICHD) Randomized Trial. JAMA 2003, 289, 3106–3116. [Google Scholar] [CrossRef] [Green Version]
  160. Taylor, R.S.; Richards, S.H.; Anderson, L.; Jenkinson, C.E.; Whalley, B.; Rees, K.; Davies, P.; Bennett, P.; Liu, Z.; West, R.; et al. Psychological interventions for coronary heart disease. Cochrane Libr. 2017, 2021, CD002902. [Google Scholar] [CrossRef]
  161. van Halewijn, G.; Deckers, J.; Tay, H.Y.; van Domburg, R.; Kotseva, K.; Wood, D. Lessons from contemporary trials of cardiovascular prevention and rehabilitation: A systematic review and meta-analysis. Int. J. Cardiol. 2016, 232, 294–303. [Google Scholar] [CrossRef] [Green Version]
  162. Blumenthal, J.A.; Sherwood, A.; Smith, P.J.; Watkins, L.; Mabe, S.; Kraus, W.E.; Ingle, K.; Miller, P.; Hinderliter, A. Enhancing Cardiac Rehabilitation with Stress Management Training: A Randomized, Clinical Efficacy Trial. Circulation 2016, 133, 1341–1350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Visseren, F.L.J.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C.; Bäck, M.; Benetos, A.; Biffi, A.; Boavida, J.-M.; Capodanno, D.; et al. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice: Developed by the Task Force for cardiovascular disease prevention in clinical practice with representatives of the European Society of Cardiology and 12 medical societies with the special contribution of the European Association of Preventive Cardiology (EAPC). Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef] [PubMed]
  164. Edmondson, D.; Newman, J.D.; Whang, W.; Davidson, K.W. Emotional triggers in myocardial infarction: Do they matter? Eur. Heart J. 2012, 34, 300–306. [Google Scholar] [CrossRef] [PubMed]
  165. Kraynak, T.E.; Marsland, A.L.; Gianaros, P.J. Neural Mechanisms Linking Emotion with Cardiovascular Disease. Curr. Cardiol. Rep. 2018, 20, 128. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic model of the mechanisms that translate negative emotion into CV disease. The brain response to mental stress involves activation of autonomic nervous system, hypothalamic–pituitary–adrenal axis, and neural–hematopoietic–arterial axis, resulting in pathophysiological effects that trigger CV events (brain-to-body). In turn, these effects modulate the brain response to stress (body-to-brain).
Figure 1. Schematic model of the mechanisms that translate negative emotion into CV disease. The brain response to mental stress involves activation of autonomic nervous system, hypothalamic–pituitary–adrenal axis, and neural–hematopoietic–arterial axis, resulting in pathophysiological effects that trigger CV events (brain-to-body). In turn, these effects modulate the brain response to stress (body-to-brain).
Jcm 11 04405 g001
Figure 2. A 73-year-old woman admitted for anterior ST-elevation myocardial infarction. Coronary angiography showed no coronary lesions on left and right coronary arteries (frames (1) and (2)). Left ventriculogram (frames (3) and (4)) showed typical ballooning of the left ventricle apex (frame (4)), characterised by hypercontraction of the basal segments (with asterisks) and akinesia in the mid and apical segments (with arrows), typical for Takotsubo cardiomyopathy.
Figure 2. A 73-year-old woman admitted for anterior ST-elevation myocardial infarction. Coronary angiography showed no coronary lesions on left and right coronary arteries (frames (1) and (2)). Left ventriculogram (frames (3) and (4)) showed typical ballooning of the left ventricle apex (frame (4)), characterised by hypercontraction of the basal segments (with asterisks) and akinesia in the mid and apical segments (with arrows), typical for Takotsubo cardiomyopathy.
Jcm 11 04405 g002
Figure 3. A 43-year-old man admitted for anterior ST-elevation myocardial infarction. Coronary angiography showed no coronary lesions on left and right coronary arteries (frames (1) and (2)). Left ventriculogram (frames (3) and (4)) showed hypercontraction of the basal segments and apex (frame (4), with asterisks) and akinesia in the mid segments (with arrows), typical for mid-ventricular Takotsubo cardiomyopathy.
Figure 3. A 43-year-old man admitted for anterior ST-elevation myocardial infarction. Coronary angiography showed no coronary lesions on left and right coronary arteries (frames (1) and (2)). Left ventriculogram (frames (3) and (4)) showed hypercontraction of the basal segments and apex (frame (4), with asterisks) and akinesia in the mid segments (with arrows), typical for mid-ventricular Takotsubo cardiomyopathy.
Jcm 11 04405 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Henein, M.Y.; Vancheri, S.; Longo, G.; Vancheri, F. The Impact of Mental Stress on Cardiovascular Health—Part II. J. Clin. Med. 2022, 11, 4405. https://doi.org/10.3390/jcm11154405

AMA Style

Henein MY, Vancheri S, Longo G, Vancheri F. The Impact of Mental Stress on Cardiovascular Health—Part II. Journal of Clinical Medicine. 2022; 11(15):4405. https://doi.org/10.3390/jcm11154405

Chicago/Turabian Style

Henein, Michael Y., Sergio Vancheri, Giovanni Longo, and Federico Vancheri. 2022. "The Impact of Mental Stress on Cardiovascular Health—Part II" Journal of Clinical Medicine 11, no. 15: 4405. https://doi.org/10.3390/jcm11154405

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop