Next Article in Journal
ppGpp, the General Stress Response Alarmone, Is Required for the Expression of the α-Hemolysin Toxin in the Uropathogenic Escherichia coli Isolate, J96
Next Article in Special Issue
Endogenous Modulation of Extracellular Matrix Collagen during Scar Formation after Myocardial Infarction
Previous Article in Journal
Proteomics Unveils Post-Mortem Changes in Beef Muscle Proteins and Provides Insight into Variations in Meat Quality Traits of Crossbred Young Steers and Heifers Raised in Feedlot
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms of Takotsubo Syndrome

1
Department of Cardiovascular Medicine, University of Oxford, Oxford OX1 2JD, UK
2
Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, 30625 Hannover, Germany
3
Fraunhofer Institute of Toxicology and Experimental Medicine, 30625 Hannover, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12262; https://doi.org/10.3390/ijms232012262
Submission received: 19 September 2022 / Revised: 7 October 2022 / Accepted: 10 October 2022 / Published: 14 October 2022
(This article belongs to the Special Issue Mechanisms of Cardiovascular Disease: Molecular Perspective 2.0)

Abstract

:
Takotsubo syndrome (TTS) is a severe but reversible acute heart failure syndrome that occurs following high catecholaminergic stress. TTS patients are similar to those with acute coronary syndrome, with chest pain, dyspnoea and ST segment changes on electrocardiogram, but are characterised by apical akinesia of the left ventricle, with basal hyperkinesia in the absence of culprit coronary artery stenosis. The pathophysiology of TTS is not completely understood and there is a paucity of evidence to guide treatment. The mechanisms of TTS are thought to involve catecholaminergic myocardial stunning, microvascular dysfunction, increased inflammation and changes in cardiomyocyte metabolism. Here, we summarise the available literature to focus on the molecular basis for the pathophysiology of TTS to advance the understanding of the condition.

1. Introduction

Takotsubo syndrome (TTS) is a severe but reversible acute heart failure syndrome that results from catecholaminergic stress [1,2,3]. TTS patients typically present with chest pain and dyspnoea, and often have ST-segment elevation on electrocardiogram (ECG) [4]. Consequently, TTS patients are often initially suspected to have acute myocardial infarction (AMI), and 1–2% of patients with suspected acute coronary syndrome (ACS) [5,6] and up to 5–6% of female patients presenting with suspected STEMI [7] are eventually diagnosed with TTS.
TTS characteristically involves apical akinesia of the left ventricle, with concomitant basal hyperkinesia. Whilst this may involve other patterns of contractile dysfunction in a minority of cases, the acute contractile dysfunction in TTS occurs in the absence of culprit coronary artery disease [4]. TTS predominantly affects post-menopausal women, and typically occurs following extreme physical or emotional stress [2,4]. Serious complications result from the extreme ventricular dilatation and reduced cardiac output, including cardiogenic shock, thrombi formation, pulmonary oedema, arrhythmia generation and LV rupture. TTS is associated with significant mortality burden, with short- and long-term mortality similar to AMI [7,8]. Patients also experience long-term contractile dysfunction [9], and are at approximately a 10% risk of recurrence [10].
Since there is no evidence-based treatment for TTS, understanding its pathophysiology is important. Here, we summarise the available literature by performing searches into TTS and the adrenergic system, microvascular dysfunction, oestrogen, genetic changes and biomarkers to produce this review focussing on the molecular mechanisms of TTS.

2. Cardiomyocyte Contractility and the Adrenergic System

Catecholamines are evidenced to play a causative role in TTS. TTS has been frequently reported to occur secondary to medical conditions with elated catecholamine levels such as pheochromocytoma [11,12,13], acute subarachnoid haemorrhage [14,15] and acute thyrotoxicosis [16]. Further, iatrogenic TTS occurs in stress dobutamine echocardiography and after adrenaline administration [17,18]. Adrenaline levels were reported by Wittstein et al. to be 10 to 20 times normal in 13 patients with TTS, and also higher than in STEMI [19]. Pre-clinical models of TTS provide additional evidence, as TTS can be robustly induced in rodent and primate models with adrenaline [20,21]. More recent studies, however, have failed to demonstrate elevated catecholamine levels in TTS patients [22], although circulating catecholamine levels are extremely short lived [23].
The central involvement of β-adrenergic receptors (βARs) was suggested by Lyon et al. to underpin the pattern of regional wall motion abnormalities (RWMA) that occur in TTS [24]. In normal physiological conditions, βARs modulate rate and force of myocardial contraction and relaxation, allowing response to stress or exercise. This process becomes dysregulated in disease, and beta-blockade is a mainstay of heart failure therapy.
βARs are spatially organized across the left ventricle (LV), with the highest density in the LV apex [20,25,26] which become exposed to circulating adrenaline. In contrast, the base of the LV has a greater density of sympathetic nerve terminals [27] and tends to receive stimulation via noradrenaline. In particular, the human myocardium demonstrates a higher concentration of β2AR when compared to other mammals (β1AR to β2AR ratio of 4:1), with minimal expression of β3AR [28,29]. Indeed, this apicobasal difference is demonstrated by larger apical βAR response in vivo [30,31,32] and β2AR response in vitro [20,33], which has been demonstrated to be consequent to fewer caveolae (sequestration) and lower cAMP buffering by PDEs [33].
Both β1AR and β2AR signal via the canonical stimulatory Gαs pathway, which leads to activation of the enzyme adenylyl cyclase (AC) and increases intracellular cAMP [34,35]. The intracellular cAMP compartmentalisation of these respective adrenoceptors differ, with β1AR-Gαs resulting in cell wide increases in cAMP whereas β2AR-Gαs results in localised signalling. cAMP subsequently activates PKA which phosphorylates downstream protein targets to result in positive inotropy, lusitropy and chronotropy.
At higher agonist concentrations, the pleiotropic β2AR can also signal via the inhibitory Gαi [36,37], and physiologically, this limits the pro-apoptotic and pro-arrhythmic toxic effects of Gαs activity [38]. This shifts receptor coupling to Gαi, in a process known as stimulus trafficking or biased agonism [36]. Signalling via cAMP-PKA phosphorylates the β2AR and primes the receptor for subsequent phosphorylation by G-protein coupled receptor kinase (GRK) whereby GRK is recruited by Gβγ [39]. This results in internalisation of the β2AR and stimulus trafficking to β2AR-Gαi in a two-step process. This directly and indirectly opposes Gαs activity and is negatively inotropic by downregulating cAMP activity and activating further signalling pathways including p38-MAPK [40].
Excess circulating adrenaline release in TTS may result in extreme negative inotropy via the β2AR-Gαi [24], which typically results in apical hypokinesia owing to the gradient of adrenergic receptors within the heart as previously discussed. This hypothesis is supported by an in silico modelling study that recreated Takotsubo-like contractile dysfunction with apical dysfunction following intense agonist stimulation when apical-basal β1AR and β2AR gradients were introduced [41]. Further, TTS-like dysfunction can be induced in vivo by bolus injection adrenaline or isoprenaline [20,21,42,43,44]. Interestingly, the hyperthermic response subsequent to catecholamine administration seems necessary for the induction of TTS, since a recent in vivo study demonstrated that maintenance of hyperthermia would induce TTS-like contractile dysfunction following isoprenaline administration, whereas active cooling prevented, but did not reverse, these changes [43].
The pivotal role of β2AR-Gαi in TTS induction has been demonstrated in vivo, with adrenaline-induced negative inotropy prevented with Gαi inhibition using pertussis toxin (PTX) in vivo and in vitro [20]. However, as in resting physiology, β2AR-Gαi appears cardioprotective in TTS, as complete inhibition with β2AR antagonists or PTX prevents apical hypokinesis but also increases mortality by approximately 70% [42]. Further, specific inhibition of the β2AR-Gαi-p38MAP kinase pathway in vivo TTS also increases mortality [20].
Changes in G-protein expression were noted with upregulation of TTS-associated microRNAs (miR-16 and miR-26a), whereby predisposition to TTS generation was noted in vivo following epinephrine administration [44]. This was associated with reductions in RGS4 (regulator of G-protein signalling 4) and G-protein subunit Gβ (GNB1); [44] proteins which are involved in the termination of β2AR-Gαi signalling in resting physiology. This potentiated Gαi activity was noted in the reduction in baseline contractility of apical but not basal rat cardiomyocytes with upregulation of TTS-associated microRNAs, and was reversed on inhibition of Gαi with PTX [44].
Nef et al. analysed endomyocardial biopsy samples from 16 patients with TTS to show increased activation of PI3K and Akt that was found to be normal at follow-up and is absent in ischemia [45]. These pathways are linked to β2AR-Gαi activity and promote cardiomyocyte survival. More recently, Nakano et al. observed proteins involved in β2AR-Gαi signalling to be increased in tissue samples from 26 patients with acute TTS, including increased levels of GRK and β-arrestin [46], proteins that are necessary for β2AR-Gαi stimulus trafficking to occur.
Changes in contractility may also occur via direct changes in cardiomyocyte calcium handing. Reduced calcium transient amplitude, SR calcium content and peak calcium current amplitude were found in apical cardiomyocytes when TTS-associated miRs were upregulated, and this was found to consequent from downregulation of CACNB1 (L-type calcium channel Cavβ subunit) [44], a channel that controls LTCC current [47]. Conversely, increased calcium transient amplitude and kinetics were observed in an induced-pluripotent stem cell derived cardiomyocytes (iPSC-CMs) line produced from TTS patients [48]. This was observed alongside increased β1AR, β2AR and cAMP responses, altered metabolism and increased intracellular lipid accumulation [48]. This TTS cell line demonstrated reduced baseline force generation when engineered heart tissue (EHT) strips of iPSC-CMs were produced, and these had increased sensitivity and reduced desensitization to catecholamines [48].

3. Vascular Dysfunction

After its identification, TTS was initially suggested to result from multivessel coronary vasospasm [49,50]. Subsequently, it has been linked to vasomotor dysfunction with increased vascular reactivity and altered endothelial function following psychological stress in patients with previous TTS [51]. Microvascular dysfunction has also been demonstrated in cases of TTS [52,53] which has the potential to better explain the phenotype of acute ischaemic stunning [54]. Early studies found myocardial contrast echocardiography demonstrated myocardial perfusion abnormality in patients with acute TTS which was partially reversible by adenosine and not present after 1 month, unlike in STEMI where the perfusion defect is fixed [55]. However, induction of TTS-like cardiac dysfunction in rat with isoprenaline produces myocardial dysfunction without preceding myocardial perfusion defects [56]. As such, vascular or microvascular dysfunction observed may be a consequence of the catecholamine surge in TTS. Interestingly, the CIRCUS-TTS study did not find any differences in systemic microvascular function between TTS, MI or healthy control both acutely or after 3 month follow-up [57].
The catecholamine surge that occurs in TTS likely leads to endothelial dysfunction [58] which could sensitise to vasospasm upon provocation. Prevalence of vascular dysfunction seems variable in TTS patients and has often been found to be absent [59,60,61]. Endothelin is increased in TTS and could mediate hypothetical vasospasm, however endothelin is increased to the same degree in STEMI [62] where the phenomenon of catecholamine-induced contractile dysfunction does not occur. Indeed, catecholamines such as adrenaline or dobutamine [17] vasodilate coronary arteries, and preclinical models inducing TTS with catecholamine in the absence endothelin cause apical dysfunction [20,42] in the absence of myocardial perfusion abnormality [56]. Given a diagnostic criterion of TTS is the presence of contractile abnormality usually occurring over more than one coronary territory [63], phenotypes of TTS would not be explained by single or multivessel spasm, and endomyocardial biopsies show absence of ischaemic, stunned or hibernating myocardium [54,64]. Since TTS is similar in presentation to ACS, it has been suggested that TTS is a form of microvascular or aborted MI [65]. Despite this, intravascular ultrasound studies in acute TTS have not found plaque rupture, endothelial breach or intracoronary thrombus [66,67], and there is no difference in incidence of wrap-around left anterior descending coronary artery in TTS [68]. Further, long-term recurrence of TTS is not influenced by aspirin therapy [69].

4. Oestrogen Deprivation

Oestrogen confers cardioprotection via a multitude of mechanisms, including sympatholysis [70,71], and 80% of TTS patients are post-menopausal women [4]. Basal circulating adrenaline is lower in women than in men [72], as is urinary cortisol, adrenaline and noradrenaline, which rise with age [73]. Interestingly, hormone replacement therapy reduced the levels of these stress hormones, suggesting regulation by oestrogen [73]. However, there is no difference in oestrogen levels between female TTS patients and age-matched patients with MI [74]. Women demonstrate greater vascular β2AR sensitivity [75] which suggests oestrogen may be able to potentiate β2AR signalling. Oestrogen can signal by downstream β2AR-Gαi-PI3K and -Akt pro-survival pathways [76] which confer cardioprotective effects [24] as earlier discussed.
However, agonist stimulation of the G protein-coupled oestrogen receptor (GPER) with G1/E2 prevented the TTS-like contractile changes elicited by high dose adrenaline in rodent [77]. In vitro, G1/E2 reduced phosphorylation and internalization of β2AR [77], and Gαi activity, and increased cAMP concentration in cardiomyocytes treated with adrenaline [77]. Further, in these experiments G1/E2 prevented the decreased Ca2+ amplitude and channel current (ICa-L) caused by adrenaline in rat cardiomyocytes [77].
Adrenergic dependent gene expression changes in immediate early genes (IEGs) in endothelial cells, myocardial cells and coronary smooth muscle cells, including c-fos but not c-jun, rapid activation of p44/p42 MAP kinase and heat shock protein 70, are observed in a rodent model of emotional stress causing TTS by conscious immobilization [78,79], and are prevented by oestrogen supplementation [80]. A potent vasodilator failed to inhibit expression of IEG mRNAs suggesting these changes are not driven by ischemia [78]. Oophorectomy promotes sympathetic activity by upregulating β1AR expression, which is reversed by subsequent reintroduction of oestrogen [81]. Further, β1AR upregulation caused by exposure to catecholamines and following ischemia-reperfusion injury is reduced by chronic oestrogen exposure [82]. The impact of hormone replacement therapy in post-menopausal women on TTS incidence and recurrence is not clear, and offers an interesting avenue for further investigation and possible prevention of TTS.

5. Genomic Changes

Efforts have been made to identify genetic mutations in patients with TTS, however these are poorly conserved and relatively few have been found. The L41Q GRK5 ‘gain-of-function’ polymorphism in transgenic mice and isolated cells results in enhanced cardiac GRK5 activity, βAR phosphorylation and βAR desensitization, and causes negative inotropy after high catecholamine release [83]. Whilst GRK5 polymorphism was found to be more common in two Italian TTS cohorts [84,85], this finding has not been demonstrated elsewhere [86]. Although examples of family predilection to TTS have been noted in several case series [87,88,89,90], specific genetic mutations have not been identified. Several functional polymorphisms have been proposed and identified in isolated cases for α1AR, β1AR, β2AR, GRK5 and oestrogen receptors [91]. Further, whole exome sequencing of the aforementioned TTS iPSC-CM patient line did not identify any genetic differences [48].
An additional source of genomic variation may exist in different disease states from non-coding RNA (ncRNA), which can be utilised both in biomarker detection [92] and in understanding disease pathophysiology. miR-1, miR-16, miR-26a and miR-133a are significantly raised in TTS patients when compared to healthy control [62]. However, the profile of these differences enabled differentiation from ST-elevation myocardial infarction (STEMI), which TTS is often confused for at patient presentation. miR-1 and miR-133a are significantly higher in STEMI whereas miR-16 and miR-26a were only significantly raised in TTS, and not STEMI [62]. These ‘TTS-associated miRs’ (miR-16 and miR-26a) have recently been identified to be involved in the pathophysiology of TTS, where they sensitise to TTS-like changes in contractility in vitro and in vivo following adrenaline, and cause TTS to be induced in vivo at lower adrenaline concentrations [44]. In full, miR-16 and/or miR-26a upregulation in isolated apical, but not basal, adult rat cardiomyocytes reduce baseline contractility [44]. This was reversed with PTX (as discussed above) and reproduced in non-failing human cardiomyocytes [44]. Sensitivity to adrenaline was reduced in apical cardiomyocytes with increased TTS-miRs, and the positive inotropic effect of adrenaline was increased only in basal cardiomyocytes [44]. These changes occurred via reduction in protein level of CACNB1 (L-type calcium channel Cavβ subunit), RGS4 (regulator of G-protein signalling 4) and G-protein subunit Gβ (GNB1) [44] as discussed above.
Mutation in Bcl2-associated athanogene 3 (BAG3) 3′-UTR (a component of the chaperone-assisted autophagy pathway) has been noted in a TTS patient cohort that prevented miR-371a-5p binding and increased level of BAG3 in cardiomyocytes following exposure to adrenaline [93]. However, this report suggests increased protein expression of BAG3 following miR-371a binding which represents an atypical mechanism of miR regulation, and it remains unclear how changes in BAG3 contribute in TTS.

6. Inflammatory Signalling

Beyond the initial contractile dysfunction present in TTS, long-term sequelae following high adrenaline appear to involve cardiac inflammation. Endomyocardial biopsy from TTS patients demonstrate mononuclear infiltrates and contraction-band necrosis [19], and slowly resolving myocardial oedema is present on cardiac magnetic resonance imaging (CMR) [94]. As the acute oedema subsides, global microscopic fibrosis develops and is detected from 4 months [9]. This occurs alongside gene changes in metabolic pathways with a shift to lipid based metabolism [95]. This is associated with symptomatic and functional impairment after greater than 1 year following diagnosis of TTS, and is associated with persistent subclinical cardiac dysfunction [96].
Presence of nitrosative stress has been identified by immunohistological studies in LV myocardium from TTS [97], and altered nitric oxide (NO) signalling has also been demonstrated [98]. In vivo induction of TTS with isoprenaline in rodent has demonstrated increased CD68+ macrophages and levels of inflammatory markers [99]. Whilst inhibition of PARP-1 (Poly [ADP-ribose] polymerase 1) partially reversed apical radial strain and fractional shortening [99], this has no effect on inflammatory markers or oxidative stress [100].
TTS patients have also been observed to have a greater retention of ultra-small iron oxide superparamagnetic particle (USPIO - phagocytosed by activated tissue macrophages) which was not detectable at 5 month follow-up, and therefore their increased activity drives the increased myocardial inflammation in TTS [101]. Interleukin-6 and chemokine (C-X-C motif) ligand 1 were increased in this context, and macrophage subtype was shifted from CD14++CD16+ and non-classic monocytes to CD14++CD16- [101]. TTS induced in rodent with isoprenaline demonstrates localised myocardial inflammatory changes followed by clusters of predominantly M1 proinflammatory macrophages, a finding seen in post-mortem myocardial samples from TTS patients [102]. Interestingly this study found M2 macrophage levels to be correlated with positive recovery of cardiac function [102]. Whilst this inflammatory change has been observed in vivo following direct administration of catecholamines [102], TTS has been triggered by immune checkpoint inhibitors which activate T lymphocytes and increase inflammation [103,104].

7. Mitochondrial Dysfunction

Alongside apical contractile dysfunction seen in rats treated with high dose catecholamines, intracellular lipid deposition is seen which supports the presence of acute mitochondrial dysfunction [42], as well as apoptosis [105] and fibrosis [106]. Similar findings have been observed in clinical TTS [64,107]. Comparison of TTS iPSC-CMs also observed increased intracellular lipid accumulation and reduced mitochondrial function [48].
Recent data from an isoprenaline-induced TTS model shows significant alterations in glucose and fatty acid metabolism as well as Krebs cycle activity in TTS [108]. Indeed, cardiac 18F-FDG metabolic rate was found to be increased in TTS in rodent, and expression of GLUT4-RNA/GLUT1/HK2-RNA was increased, with accumulation of glucose- and fructose-6-phosphates and increased hexokinase activity [108]. There was a shift from lactate and pyruvate to β-Oxidation enzymes CPT1b-RNA and 3-ketoacyl-CoA thiolase, and although malonyl-CoA (CPT-1 regulator) activity was seen to be increased, fatty acids and acyl-carnitines levels were reduced [108]. Krebs cycle intermediates α-ketoglutarate and succinyl-carnitine, along with dihydroorotate (a cellular ATP reporter) were reduced [108]. Mitochondrial Ca2+ uptake was initially impaired, inducing oxidation of NAD(P)H and FAD [108].

8. Brain Heart Axis

TTS was shown to be closely associated with neuropsychiatric disorders by Templin et al. from their study of 1750 patients with TTS [4], and there seems to be a clear interaction between brain and heart in this context. Whilst original data looked at various neurological and psychiatric conditions [4], more recent literature review suggested that pre-admission anxiety disorders were the most prevalent abnormalities in patients presenting with acute TTS [109]. TTS-associated miRs recently [62] have also been shown to be altered in neuropsychiatric stress [110,111,112]. Consequently it is interesting that they are involved in the predisposition to TTS generation in rodent as above [44], and this suggests that chronic neuropsychiatric stress may predispose to future generation of TTS.
MRI studies have assessed the role of brain regions associated with the autonomic nervous system (ANS) and emotion, with altered brain architecture being observed in patients following TTS. Specifically, reduced insula and cingulate cortex thickness has been noted, alongside reduced connectivity in the limbic system and ANS-specific network [113]. This included alterations in the left amygdala, hippocampi, left para-hippocampal gyrus, left superior temporal pole, and right putamen [113]. Both parasympathetic and sympathetic systems are altered, suggesting the balance within the ANS may be more important than the sympathetic activity alone [114]. Indeed, there is hypoconnectivity of central brain regions associated with autonomic functions and regulation of the limbic system in patients with TTS [115]. Whether this is a cause or consequence of the large catecholamine burden experienced by TTS patients is unknown. This could explain the ‘gain’ in the hypothalamic-pituitary axis as previously suggested [2,19].
An interesting study that retrospectively identified individuals who underwent clinical 18 F-FDG-PET/CT imaging prior to generation of TTS found that TTS patient had higher baseline amygdalar activity, and amygdalar activity was associated with the risk of subsequent TTS generation [116]. Further, TTS patients with higher amygdalar activity were likely to develop TTS approximately 2 years sooner that those with low activity [116].
Neurogenic stunned myocardium (NSM) represents a condition that demonstrates characteristic myocardial contractile dysfunction following increased sympathetic activity after neurological insult. It has therefore been compared to TTS [117,118,119,120]. Whilst NSM is observed to have higher catecholamines levels, echocardiographic findings are similar to TTS [119]. Although there are minor differences in presentation [121], NSM could be a specific subset of TTS consequent from increased neurological sympathetic activity as opposed to the classical increase in circulating catecholamine that occurs in TTS. The conditions have previously been compared in great detail to conflicting opinions [119,120,122].

9. Biomarkers

Owing to the modest ischaemic damage and profound contractile abnormality present in TTS, modest increases in creatine kinase-MB and cardiac troponin are seen when compared to AMI, versus the significant elevation of natriuretic peptides, including brain natriuretic peptide (BNP) or N-terminal-pro-BNP (NT-pro-BNP) [123,124,125]. Indeed, admission NT-pro-BNP levels have been shown to act as an independent predictor of morbidity and mortality [126].
TTS-associated miRs have been discussed above, whereby miR-1, miR-16, miR-26a and miR-133a are increased in TTS compared to healthy patients, but miR-1 and miR-133a are higher in STEMI than TTS, whereas miR-16 and miR-26a were raised only in TTS [62]. The potential importance that biomarkers play in understanding the pathophysiology of TTS was illustrated in a follow-up study, showing miR-16 and miR-26a were involved in the predisposition to TTS generation in rodent as above [44].
Given the previously discussed involvement of inflammation and microvascular dysfunction in TTS [1,3], markers for these have been investigated for use as biomarkers. IL-2, IL-4, IL-10, IFN-γ and TNF-α have been seen to be higher in acute TTS than in AMI, with IL-6 being higher in AMI [127]. Further, co-peptin and endothelin, two vasocontricting peptides, are altered in TTS when compared to AMI. Co-peptin levels are substantially increased in AMI but normal or only modestly elevated in TTS [128,129,130]. When compared to control patients, endothelin levels are increased to a similar level in TTS and STEMI [62], and glycocalyx levels also seem elevated in TTS [131].

10. Conclusions

The molecular mechanisms of TTS are becoming further elucidated. There is a clear pathogenic link to a catecholamine surge which causes changes in cardiomyocyte contractility through the beta-adrenergic system. Oestrogen withdrawal post-menopause plays a role in predisposing patients to developing TTS, and it seems that chronic stress may also increase the future likelihood of TTS following acute stress. Circulating adrenaline also acts on the non-myocyte cellular population within the heart which likely results in microvascular dysfunction, however it is unclear if this contributes to contractile dysfunction. Further mechanisms include changes in cardiomyocyte metabolism and persistent inflammation which likely contributes to long-term cardiac dysfunction.

Funding

T.T. is supported this review through DFG (KFO311).

Conflicts of Interest

T.T. filed and licensed patents in the field of noncoding RNAs. T.T. is founder and shareholder of Cardior Pharmaceuticals GmbH (Hannover, Germany).

References

  1. Omerovic, E.; Citro, R.; Bossone, E.; Redfors, B.; Backs, J.; Bruns, B.; Ciccarelli, M.; Couch, L.S.; Dawson, D.; Grassi, G.; et al. Pathophysiology of Takotsubo syndrome—A joint scientific statement from the HFA TTS Study Group and Myocardial Function Working Group of the ESC—Part 1: Overview and the central role for catecholamines and sympathetic nervous system. Eur. J. Heart Fail. 2022, 24, 257–273. [Google Scholar] [CrossRef]
  2. Lyon, A.R.; Bossone, E.; Schneider, B.; Sechtem, U.; Citro, R.; Underwood, S.R.; Sheppard, M.N.; Figtree, G.A.; Parodi, G.; Akashi, Y.J.; et al. Current state of knowledge on Takotsubo syndrome: A Position Statement from the Taskforce on Takotsubo Syndrome of the Heart Failure Association of the European Society of Cardiology. Eur. J. Heart Fail 2016, 18, 8–27. [Google Scholar] [CrossRef] [Green Version]
  3. Omerovic, E.; Citro, R.; Bossone, E.; Redfors, B.; Backs, J.; Bruns, B.; Ciccarelli, M.; Couch, L.S.; Dawson, D.; Grassi, G.; et al. Pathophysiology of Takotsubo syndrome—A joint scientific statement from the HFA TTS and Myocardial Function Working Group of the ESC—Part 2: Vascular pathophysiology, gender and sex hormones, genetics, chronic cardiovascular problems and clinical impl. Eur. J. Heart Fail. 2022, 24, 274–286. [Google Scholar] [CrossRef]
  4. 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] [Green Version]
  5. Gianni, M.; Dentali, F.; Grandi, A.M.; Sumner, G.; Hiralal, R.; Lonn, E. Apical ballooning syndrome or takotsubo cardiomyopathy: A systematic review. Eur. Heart J. 2006, 27, 1523–1529. [Google Scholar] [CrossRef] [Green Version]
  6. Kurowski, V.; Kaiser, A.; Von Hof, K.; Killermann, D.P.; Mayer, B.; Hartmann, F.; Schunkert, H.; Radke, P.W. Apical and midventricular transient left ventricular dysfunction syndrome (tako-tsubo cardiomyopathy): Frequency, mechanisms, and prognosis. Chest 2007, 132, 809–816. [Google Scholar] [CrossRef]
  7. Redfors, B.; Vedad, R.; Angerås, O.; Råmunddal, T.; Petursson, P.; Haraldsson, I.; Ali, A.; Dworeck, C.; Odenstedt, J.; Ioaness, D.; et al. Mortality in takotsubo syndrome is similar to mortality in myocardial infarction—A report from the SWEDEHEART. Int. J. Cardiol. 2015, 185, 282–289. [Google Scholar] [CrossRef]
  8. Ghadri, J.R.; Kato, K.; Cammann, V.L.; Gili, S.; Jurisic, S.; Di Vece, D.; Candreva, A.; Ding, K.J.; Micek, J.; Szawan, K.A.; et al. Long-Term Prognosis of Patients With Takotsubo Syndrome. J. Am. Coll. Cardiol. 2018, 72, 874–882. [Google Scholar] [CrossRef]
  9. Schwarz, K.; Ahearn, T.; Srinivasan, J.; Neil, C.J.; Scally, C.; Rudd, A.; Jagpal, B.; Frenneaux, M.P.; Pislaru, C.; Horowitz, J.D.; et al. Alterations in Cardiac Deformation, Timing of Contraction and Relaxation, and Early Myocardial Fibrosis Accompany the Apparent Recovery of Acute Stress-Induced (Takotsubo) Cardiomyopathy: An End to the Concept of Transience. J. Am. Soc. Echocardiogr. 2017, 30, 745–755. [Google Scholar] [CrossRef] [Green Version]
  10. Kato, K.; Di Vece, D.; Cammann, V.L.; Micek, J.; Szawan, K.A.; Bacchi, B.; Lüscher, T.F.; Ruschitzka, F.; Ghadri, J.R.; Templin, C. Takotsubo Recurrence: Morphological Types and Triggers and Identification of Risk Factors. J. Am. Coll. Cardiol. 2019, 73, 982–984. [Google Scholar] [CrossRef]
  11. Y-Hassan, S. Clinical Features and Outcome of Pheochromocytoma-Induced Takotsubo Syndrome: Analysis of 80 Published Cases. Am. J. Cardiol. 2016, 117, 1836–1844. [Google Scholar] [CrossRef] [PubMed]
  12. Prejbisz, A.; Lenders, J.W.; Eisenhofer, G.; Januszewicz, A. Cardiovascular manifestations of phaeochromocytoma. J. Hypertens. 2011, 29, 2049–2060. [Google Scholar] [CrossRef] [PubMed]
  13. Agarwal, V.; Kant, G.; Hans, N.; Messerli, F.H. Takotsubo-like cardiomyopathy in pheochromocytoma. Int. J. Cardiol. 2011, 153, 241–248. [Google Scholar] [CrossRef] [PubMed]
  14. Naredi, S.; Lambert, G.; Edén, E.; Zäll, S.; Runnerstam, M.; Rydenhag, B.; Friberg, P. Increased sympathetic nervous activity in patients with nontraumatic subarachnoid hemorrhage. Stroke 2000, 31, 901–906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Redfors, B.; Shao, Y.; Omerovic, E. Stress-induced cardiomyopathy (Takotsubo)—Broken heart and mind? Vasc. Health Risk Manag. 2013, 9, 149–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Eliades, M.; El-Maouche, D.; Choudhary, C.; Zinsmeister, B.; Burman, K.D. Takotsubo cardiomyopathy associated with thyrotoxicosis: A case report and review of the literature. Thyroid 2014, 24, 383–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Abraham, J.; Mudd, J.O.; Kapur, N.; Klein, K.; Champion, H.C.; Wittstein, I.S. Stress Cardiomyopathy After Intravenous Administration of Catecholamines and Beta-Receptor Agonists. J. Am. Coll. Cardiol. 2009, 53, 1320–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Redfors, B.; Shao, Y.; Omerovic, E. Stress-induced cardiomyopathy in a patient with chronic spinal cord transection at the level of C5: Endocrinologically mediated catecholamine toxicity. Int. J. Cardiol. 2012, 159, e61–e62. [Google Scholar] [CrossRef] [PubMed]
  19. Wittstein, I.S.; Thiemann, D.R.; Lima, J.; Baughman, K.L.; Schulman, S.P.; Gerstenblith, G.; Wu, K.C.; Rade, J.J.; Bivalacqua, T.J.; Champion, H.C.; et al. Neurohumoral features of myocardial stunning due to sudden emotional stress. N. Engl. J. Med. 2005, 352, 539–548. [Google Scholar] [CrossRef] [PubMed]
  20. Paur, H.; Wright, P.T.; Sikkel, M.B.; Tranter, M.H.; Mansfield, C.; O’Gara, P.; Stuckey, D.J.; Nikolaev, V.O.; Diakonov, I.; Pannell, L.; et al. High levels of circulating epinephrine trigger apical cardiodepression in a β 2-adrenergic receptor/Gi-dependent manner: A new model of takotsubo cardiomyopathy. Circulation 2012, 126, 697–706. [Google Scholar] [CrossRef] [PubMed]
  21. Izumi, Y.; Okatani, H.; Shiota, M.; Nakao, T.; Ise, R.; Kito, G.; Miura, K.; Iwao, H. Effects of metoprolol on epinephrine-induced takotsubo-like left ventricular dysfunction in non-human primates. Hypertens. Res. 2009, 32, 339–346. [Google Scholar] [CrossRef] [PubMed]
  22. Y-Hassan, S.; Sörensson, P.; Ekenbäck, C.; Lundin, M.; Agewall, S.; Brolin, E.B.; Caidahl, K.; Cederlund, K.; Collste, O.; Daniel, M.; et al. Plasma catecholamine levels in the acute and subacute stages of takotsubo syndrome: Results from the Stockholm myocardial infarction with normal coronaries 2 study. Clin. Cardiol. 2021, 44, 1567–1574. [Google Scholar] [CrossRef] [PubMed]
  23. EMC Adrenaline (Epinephrine) Injection BP 1 in 1000—Summary of Product Characteristics (SmPC) 2022. Available online: https://www.medicines.org.uk/emc/product/6284/#gref (accessed on 19 September 2022).
  24. Lyon, A.R.; Rees, P.S.C.; Prasad, S.; Poole-Wilson, P.A.; Harding, S.E. Stress (Takotsubo) cardiomyopathy—A novel pathophysiological hypothesis to explain catecholamine-induced acute myocardial stunning. Nat. Clin. Pract. Cardiovasc. Med. 2008, 5, 22–29. [Google Scholar] [CrossRef] [PubMed]
  25. Mori, H.; Ishikawa, S.; Kojima, S.; Hayashi, J.; Watanabe, Y.; Hoffman, J.I.E.; Okino, H. Increased responsiveness of left ventricular apical myocardium to adrenergic stimuli. Cardiovasc. Res. 1993, 27, 192–198. [Google Scholar] [CrossRef] [PubMed]
  26. Brouri, F.; Hanoun, N.; Mediani, O.; Saurini, F.; Hamon, M.; Vanhoutte, P.M.; Lechat, P. Blockade of beta 1- and desensitization of beta 2-adrenoceptors reduce isoprenaline-induced cardiac fibrosis. Eur. J. Pharmacol. 2004, 485, 227–234. [Google Scholar] [CrossRef] [PubMed]
  27. Kawano, H.; Okada, R.; Yano, K. Histological study on the distribution of autonomic nerves in the human heart. Heart Vessels 2003, 18, 32–39. [Google Scholar] [CrossRef] [PubMed]
  28. Bristow, M.R.; Ginsburg, R.; Umans, V.; Fowler, M.; Minobe, W.; Rasmussen, R.; Zera, P.; Menlove, R.; Shah, P.; Jamieson, S. β1- and β2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: Coupling of both receptor subtypes to muscle contraction and selective β1-receptor down-regulation in heart failure. Circ. Res. 1986, 59, 297–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Moniotte, S.; Kobzik, L.; Feron, O.; Trochu, J.N.; Gauthier, C.; Balligand, J.L. Upregulation of β3-adrenoceptors and altered contractile response to inotropic amines in human failing myocardium. Circulation 2001, 103, 1649–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Heather, L.C.; Catchpole, A.F.; Stuckey, D.J.; Cole, M.A.; Carr, C.A.; Clarke, K. Isoproterenol induces in vivo functional and metabolic abnormalities: Similar to those found in the infarcted rat heart. J. Physiol. Pharmacol. 2009, 60, 31–39. [Google Scholar] [PubMed]
  31. Mantravadi, R.; Gabris, B.; Liu, T.; Choi, B.-R.; de Groat, W.C.; Ng, G.A.; Salama, G. Autonomic nerve stimulation reverses ventricular repolarization sequence in rabbit hearts. Circ. Res. 2007, 100, e72–e80. [Google Scholar] [CrossRef] [PubMed]
  32. Lathers, C.M.; Levin, R.M.; Spivey, W.H. Regional distribution of myocardial β-adrenoceptors in the cat. Eur. J. Pharmacol. 1986, 130, 111–117. [Google Scholar] [CrossRef]
  33. Wright, P.T.; Bhogal, N.K.; Diakonov, I.; Pannell, L.M.K.; Perera, R.K.; Bork, N.I.; Schobesberger, S.; Lucarelli, C.; Faggian, G.; Alvarez-Laviada, A.; et al. Cardiomyocyte Membrane Structure and cAMP Compartmentation Produce Anatomical Variation in β2AR-cAMP Responsiveness in Murine Hearts. Cell Rep. 2018, 23, 459–469. [Google Scholar] [CrossRef] [Green Version]
  34. Rosenbaum, D.M.; Rasmussen, S.G.F.; Kobilka, B.K. The structure and function of G-protein-coupled receptors. Nature 2009, 459, 356–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Evans, B.; Sato, M.; Sarwar, M.; Hutchinson, D.; Summers, R. Ligand-directed signalling at β-adrenoceptors. Br. J. Pharmacol. 2010, 159, 1022–1038. [Google Scholar] [CrossRef] [Green Version]
  36. Heubach, J.F.; Ravens, U.; Kaumann, A.J. Epinephrine activates both Gs and Gi pathways, but norepinephrine activates only the Gs pathway through human beta2-adrenoceptors overexpressed in mouse heart. Mol. Pharmacol. 2004, 65, 1313–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hasseldine, A.R.G.; Harper, E.A.; Black, J.W. Cardiac-specific overexpression of human beta2 adrenoceptors in mice exposes coupling to both Gs and Gi proteins. Br. J. Pharmacol. 2003, 138, 1358–1366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zhu, W.; Wang, S.; Chakir, K.; Yang, D.; Zhang, T.; Brown, J.H.; Devic, E.; Kobilka, B.K.; Cheng, H.; Xiao, R.-P. Linkage of β1-adrenergic stimulation to apoptotic heart cell death through protein kinase A—Independent activation of Ca2+/calmodulin kinase II. J. Clin. 2003, 111, 617–625. [Google Scholar] [CrossRef] [Green Version]
  39. Liu, R.; Ramani, B.; Soto, D.; De Arcangelis, V.; Xiang, Y. Agonist dose-dependent phosphorylation by protein kinase A and G protein-coupled receptor kinase regulates β2 adrenoceptor coupling to Gi proteins in cardiomyocytes. J. Biol. Chem. 2009, 284, 32279–32287. [Google Scholar] [CrossRef] [Green Version]
  40. Heubach, J.F.; Blaschke, M.; Harding, S.E.; Ravens, U.; Kaumann, A.J. Cardiostimulant and cardiodepressant effects through overexpressed human β2-adrenoceptors in murine heart: Regional differences and functional role of β1-adrenoceptors. Naunyn Schmiedebergs Arch. Pharmacol. 2003, 367, 380–390. [Google Scholar] [CrossRef] [PubMed]
  41. Land, S.; Niederer, S.A.; Louch, W.E.; Røe, A.T.; Aronsen, J.M.; Stuckey, D.J.; Sikkel, M.B.; Tranter, M.H.; Lyon, A.R.; Harding, S.E.; et al. Computational modeling of Takotsubo cardiomyopathy: Effect of spatially varying β-adrenergic stimulation in the rat left ventricle. Am. J. Physiol. Heart Circ. Physiol. 2014, 307, H1487–H1496. [Google Scholar] [CrossRef]
  42. Shao, Y.; Redfors, B.; Scharin Tang, M.; Mollmann, H.; Troidl, C.; Szardien, S.; Hamm, C.; Nef, H.; Boren, J.; Omerovic, E.; et al. Novel rat model reveals important roles of β-adrenoreceptors in stress-induced cardiomyopathy. Int. J. Cardiol. 2013, 168, 1943–1950. [Google Scholar] [CrossRef] [PubMed]
  43. Tranter, M.H.; Redfors, B.; Wright, P.T.; Couch, L.S.; Lyon, A.R.; Omerovic, E.; Harding, S.E. Hyperthermia as a trigger for Takotsubo syndrome in a rat model. Front. Cardiovasc. Med. 2022, 9, 869585. [Google Scholar] [CrossRef] [PubMed]
  44. Couch, L.S.; Fiedler, J.; Chick, G.; Clayton, R.; Dries, E.; Wienecke, L.M.; Fu, L.; Fourre, J.; Pandey, P.; Derda, A.A.; et al. Circulating microRNAs predispose to takotsubo syndrome following high-dose adrenaline exposure. Cardiovasc. Res. 2022, 118, 1758–1770. [Google Scholar] [CrossRef]
  45. Nef, H.M.; Möllmann, H.; Hilpert, P.; Troidl, C.; Voss, S.; Rolf, A.; Behrens, C.B.; Weber, M.; Hamm, C.W.; Elsässer, A. Activated cell survival cascade protects cardiomyocytes from cell death in Tako-Tsubo cardiomyopathy. Eur. J. Heart Fail. 2009, 11, 758–764. [Google Scholar] [CrossRef] [PubMed]
  46. Nakano, T.; Onoue, K.; Nakada, Y.; Nakagawa, H.; Kumazawa, T.; Ueda, T.; Nishida, T.; Soeda, T.; Okayama, S.; Watanabe, M.; et al. Alteration of β-Adrenoceptor Signaling in Left Ventricle of Acute Phase Takotsubo Syndrome: A Human Study. Sci. Rep. 2018, 8, 12731. [Google Scholar] [CrossRef] [Green Version]
  47. Weissgerber, P.; Held, B.; Bloch, W.; Kaestner, L.; Chien, K.R.; Fleischmann, B.K.; Lipp, P.; Flockerzi, V.; Freichel, M. Reduced cardiac L-type Ca2+ current in Cavβ2−/− embryos impairs cardiac development and contraction with secondary defects in vascular maturation. Circ. Res. 2006, 99, 749–757. [Google Scholar] [CrossRef] [Green Version]
  48. Borchert, T.; Hübscher, D.; Guessoum, C.I.; Lam, T.-D.D.; Ghadri, J.R.; Schellinger, I.N.; Tiburcy, M.; Liaw, N.Y.; Li, Y.; Haas, J.; et al. Catecholamine-Dependent β-Adrenergic Signaling in a Pluripotent Stem Cell Model of Takotsubo Cardiomyopathy. J. Am. Coll. Cardiol. 2017, 70, 975–991. [Google Scholar] [CrossRef] [PubMed]
  49. Sato, H.; Tateishi, H.; Uchida, T.; Al, E. Takotsubo-type cardiomyopathy due to multivessel spasm. In Clinical Aspect of Myocardial Injury: From Ischemia to Heart Failure; Kodama, K., Haze, K., Hon, M., Eds.; Kagakuhyouronsha: Tokyo, Japan, 1990; pp. 56–64. [Google Scholar]
  50. Dote, K.; Sato, H.; Tateishi, H.; Uchida, T.; Ishihara, M. Myocardial stunning due to simultaneous multivessel coronary spasms: A review of 5 cases. J. Cardiol. 1991, 21, 203–214. [Google Scholar]
  51. Martin, E.A.; Prasad, A.; Rihal, C.S.; Lerman, L.O.; Lerman, A. Endothelial Function and Vascular Response to Mental Stress Are Impaired in Patients With Apical Ballooning Syndrome. J. Am. Coll. Cardiol. 2010, 56, 1840–1846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Patel, S.M.; Lerman, A.; Lennon, R.J.; Prasad, A. Impaired coronary microvascular reactivity in women with apical ballooning syndrome (Takotsubo/stress cardiomyopathy). Eur. Hear. J. Acute Cardiovasc. Care 2013, 2, 147–152. [Google Scholar] [CrossRef]
  53. Kurisu, S.; Inoue, I.; Kawagoe, T.; Ishihara, M.; Shimatani, Y.; Nishioka, K.; Umemura, T.; Nakamura, S.; Yoshida, M.; Sato, H. Myocardial perfusion and fatty acid metabolism in patients with tako-tsubo-like left ventricular dysfunction. J. Am. Coll. Cardiol. 2003, 41, 743–748. [Google Scholar] [CrossRef]
  54. Kato, K.; Lyon, A.R.; Ghadri, J.R.; Templin, C. Takotsubo syndrome: Aetiology, presentation and treatment. Heart 2017, 103, 1461–1469. [Google Scholar] [CrossRef] [PubMed]
  55. Galiuto, L.; De Caterina, A.R.; Porfidia, A.; Paraggio, L.; Barchetta, S.; Locorotondo, G.; Rebuzzi, A.G.; Crea, F. Reversible coronary microvascular dysfunction: A common pathogenetic mechanism in Apical Ballooning or Tako-Tsubo Syndrome. Eur. Heart J. 2010, 31, 1319–1327. [Google Scholar] [CrossRef] [Green Version]
  56. Redfors, B.; Shao, Y.; Wikström, J.; Lyon, A.R.; Oldfors, A.; Gan, L.M.; Omerovic, E. Contrast echocardiography reveals apparently normal coronary perfusion in a rat model of stress-induced (Takotsubo) cardiomyopathy. Eur. Heart J. Cardiovasc. Imaging 2014, 15, 152–157. [Google Scholar] [CrossRef] [Green Version]
  57. Möller, C.; Stiermaier, T.; Meusel, M.; Jung, C.; Graf, T.; Eitel, I. Microcirculation in patients with takotsubo syndrome—The prospective circus-tts study. J. Clin. Med. 2021, 10, 2127. [Google Scholar] [CrossRef]
  58. Naegele, M.; Flammer, A.J.; Enseleit, F.; Roas, S.; Frank, M.; Hirt, A.; Kaiser, P.; Cantatore, S.; Templin, C.; Fröhlich, G.; et al. Endothelial function and sympathetic nervous system activity in patients with Takotsubo syndrome. Int. J. Cardiol. 2016, 224, 226–230. [Google Scholar] [CrossRef]
  59. 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]
  60. Tsuchihashi, K.; Ueshima, K.; Uchida, T.; Oh-mura, N.; Kimura, K.; Owa, M.; Yoshiyama, M.; Miyazaki, S.; Haze, K.; Ogawa, H.; et al. Transient left ventricular apical ballooning without coronary artery stenosis: A novel heart syndrome mimicking acute myocardial infarction. Angina Pectoris-Myocardial Infarction Investigations in Japan. J. Am. Coll. Cardiol. 2001, 38, 11–18. [Google Scholar] [CrossRef] [Green Version]
  61. Abe, Y.; Kondo, M.; Matsuoka, R.; Araki, M.; Dohyama, K.; Tanio, H. Assessment of clinical features in transient left ventricular apical ballooning. J. Am. Coll. Cardiol. 2003, 41, 737–742. [Google Scholar] [CrossRef] [Green Version]
  62. Jaguszewski, M.; Osipova, J.; Ghadri, J.R.; Napp, L.C.; Widera, C.; Franke, J.; Fijalkowski, M.; Nowak, R.; Fijalkowska, M.; Volkmann, I.; et al. A signature of circulating microRNAs differentiates takotsubo cardiomyopathy from acute myocardial infarction. Eur. Heart J. 2014, 35, 999–1006. [Google Scholar] [CrossRef] [Green Version]
  63. 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]
  64. Nef, H.M.; Möllmann, H.; Kostin, S.; Troidl, C.; Voss, S.; Weber, M.; Dill, T.; Rolf, A.; Brandt, R.; Hamm, C.W.; et al. Tako-Tsubo cardiomyopathy: Intraindividual structural analysis in the acute phase and after functional recovery. Eur. Heart J. 2007, 28, 2456–2464. [Google Scholar] [CrossRef] [Green Version]
  65. 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]
  66. Delgado, G.A.; Truesdell, A.G.; Kirchner, R.M.; Zuzek, R.W.; Pomerantsev, E.V.; Gordon, P.C.; Regnante, R.A. An angiographic and intravascular ultrasound study of the left anterior descending coronary artery in takotsubo cardiomyopathy. Am. J. Cardiol. 2011, 108, 888–891. [Google Scholar] [CrossRef]
  67. Haghi, D.; Roehm, S.; Hamm, K.; Harder, N.; Suselbeck, T.; Borggrefe, M.; Papavassiliu, T. Takotsubo cardiomyopathy is not due to plaque rupture: An intravascular ultrasound study. Clin. Cardiol. 2010, 33, 307–310. [Google Scholar] [CrossRef]
  68. Hoyt, J.; Lerman, A.; Lennon, R.J.; Rihal, C.S.; Prasad, A. Left anterior descending artery length and coronary atherosclerosis in apical ballooning syndrome (Takotsubo/stress induced cardiomyopathy). Int. J. Cardiol. 2010, 145, 112–115. [Google Scholar] [CrossRef]
  69. D’Ascenzo, F.; Gili, S.; Bertaina, M.; Iannaccone, M.; Cammann, V.L.; Di Vece, D.; Kato, K.; Saglietto, A.; Szawan, K.A.; Frangieh, A.H.; et al. Impact of aspirin on takotsubo syndrome: A propensity score-based analysis of the InterTAK Registry. Eur. J. Heart Fail. 2020, 22, 330–337. [Google Scholar] [CrossRef]
  70. Ling, S.; Komesaroff, P.; Sudhir, K. Cellular mechanisms underlying the cardiovascular actions of oestrogens. Clin. Sci. 2006, 111, 107–118. [Google Scholar] [CrossRef] [Green Version]
  71. Patten, R.D.; Pourati, I.; Aronovitz, M.J.; Baur, J.; Celestin, F.; Chen, X.; Michael, A.; Haq, S.; Nuedling, S.; Grohe, C.; et al. 17β-estradiol reduces cardiomyocyte apoptosis in vivo and in vitro via activation of phospho-inositide-3 kinase/Akt signaling. Circ. Res. 2004, 95, 692–699. [Google Scholar] [CrossRef] [Green Version]
  72. Davidson, L.; Vandongen, R.; Rouse, I.L.; Beilin, L.J.; Tunney, A. Sex-related differences in resting and stimulated plasma noradrenaline and adrenaline. Clin. Sci. 1984, 67, 347–352. [Google Scholar] [CrossRef]
  73. Deane, R.; Chummun, H.; Prashad, D. Differences in urinary stress hormones in male and female nurses at different ages. J. Adv. Nurs. 2002, 37, 304–310. [Google Scholar] [CrossRef]
  74. Moller, C.; Stiermaier, T.; Brabant, G.; Graf, T.; Thiele, H.; Eitel, I. Comprehensive assessment of sex hormones in Takotsubo syndrome. Int. J. Cardiol. 2018, 250, 11–15. [Google Scholar] [CrossRef]
  75. Kneale, B.J.; Chowienczyk, P.J.; Brett, S.E.; Coltart, D.J.; Ritter, J.M. Gender differences in sensitivity to adrenergic agonists of forearm resistance vasculature. J. Am. Coll. Cardiol. 2000, 36, 1233–1238. [Google Scholar] [CrossRef] [Green Version]
  76. Deschamps, A.M.; Murphy, E. Activation of a novel estrogen receptor, GPER, is cardioprotective in male and female rats. Am. J. Physiol.-Hear. Circ. Physiol. 2009, 297, H1806–H1813. [Google Scholar] [CrossRef] [Green Version]
  77. Fu, L.; Zhang, H.; Ong’achwa Machuki, J.; Zhang, T.; Han, L.; Sang, L.; Wu, L.; Zhao, Z.; James Turley, M.; Hu, X.; et al. GPER mediates estrogen cardioprotection against epinephrine-induced stress. J. Endocrinol. 2021, 249, 209–222. [Google Scholar] [CrossRef]
  78. Ueyama, T.; Senba, E.; Kasamatsu, K.; Hano, T.; Yamamoto, K.; Nishio, I.; Tsuruo, Y.; Yoshida, K.I. Molecular mechanism of emotional stress-induced and catecholamine-induced heart attack. J. Cardiovasc. Pharmacol. 2003, 41, S115–S118. [Google Scholar]
  79. Ueyama, T.; Yoshida, K.I.; Senba, E. Emotional stress induces immediate-early gene expression in rat heart via activation of α- and β-adrenoceptors. Am. J. Physiol.-Hear. Circ. Physiol. 1999, 277, H1553–H1561. [Google Scholar] [CrossRef]
  80. Ueyama, T.; Ishikura, F.; Matsuda, A.; Asanuma, T.; Ueda, K.; Ichinose, M.; Kasamatsu, K.; Hano, T.; Akasaka, T.; Tsuruo, Y.; et al. Chronic estrogen supplementation following ovariectomy improves the emotional stress-induced cardiovascular responses by indirect action on the nervous system and by direct action on the heart. Circ. J. 2007, 71, 565–573. [Google Scholar] [CrossRef] [Green Version]
  81. Chu, S.H.; Goldspink, P.; Kowalski, J.; Beck, J.; Schwertz, D.W. Effect of estrogen on calcium-handling proteins, β-adrenergic receptors, and function in rat heart. Life Sci. 2006, 79, 1257–1267. [Google Scholar] [CrossRef]
  82. Kam, K.W.L.; Qi, J.S.; Chen, M.; Wong, T.M. Estrogen Reduces Cardiac Injury and Expression of β 1-Adrenoceptor upon Ischemic Insult in the Rat Heart. J. Pharmacol. Exp. Ther. 2004, 309, 8–15. [Google Scholar] [CrossRef] [Green Version]
  83. Liggett, S.B.; Cresci, S.; Kelly, R.J.; Syed, F.M.; Matkovich, S.J.; Hahn, H.S.; Diwan, A.; Martini, J.S.; Sparks, L.; Parekh, R.R.; et al. A GRK5 polymorphism that inhibits β-adrenergic receptor signaling is protective in heart failure. Nat. Med. 2008, 14, 510–517. [Google Scholar] [CrossRef] [Green Version]
  84. Spinelli, L.; Trimarco, V.; Di Marino, S.; Marino, M.; Iaccarino, G.; Trimarco, B. L41Q polymorphism of the G protein coupled receptor kinase 5 is associated with left ventricular apical ballooning syndrome. Eur. J. Hear. Fail. 2010, 12, 13–16. [Google Scholar] [CrossRef]
  85. Novo, G.; Giambanco, S.; Guglielmo, M.; Arvigo, L.; Sutera, M.R.; Giambanco, F.; Evola, S.; Vaccarino, L.; Bova, M.; Lio, D.; et al. G-protein-coupled receptor kinase 5 polymorphism and Takotsubo cardiomyopathy. J. Cardiovasc. Med. 2015, 16, 639–643. [Google Scholar] [CrossRef] [Green Version]
  86. Figtree, G.A.; Bagnall, R.D.; Abdulla, I.; Buchholz, S.; Galougahi, K.K.; Yan, W.; Tan, T.; Neil, C.; Horowitz, J.D.; Semsarian, C.; et al. No association of G-protein-coupled receptor kinase 5 or beta-adrenergic receptor polymorphisms with Takotsubo cardiomyopathy in a large Australian cohort. Eur. J. Hear. Fail. 2013, 15, 730–733. [Google Scholar] [CrossRef]
  87. Kumar, G.; Holmes, D.R.; Prasad, A. “Familial” apical ballooning syndrome (Takotsubo cardiomyopathy). Int. J. Cardiol. 2010, 144, 444–445. [Google Scholar] [CrossRef]
  88. Pison, L.; De Vusser, P.; Mullens, W. Apical ballooning in relatives. Heart 2004, 90, e67. [Google Scholar] [CrossRef] [PubMed]
  89. Musumeci, B.; Saponaro, A.; Pagannone, E.; Proietti, G.; Mastromarino, V.; Conti, E.; Tubaro, M.; Volpe, M.; Autore, C. Simultaneous Takotsubo syndrome in two sisters. Int. J. Cardiol. 2013, 165, e49–e50. [Google Scholar] [CrossRef] [PubMed]
  90. Ikutomi, M.; Yamasaki, M.; Matsusita, M.; Watari, Y.; Arashi, H.; Endo, G.; Yamaguchi, J.I.; Ohnishi, S. Takotsubo cardiomyopathy in siblings. Heart Vessels 2014, 29, 119–122. [Google Scholar] [CrossRef]
  91. Limongelli, G.; Masarone, D.; Maddaloni, V.; Rubino, M.; Fratta, F.; Cirillo, A.; Ludovica, S.B.; Pacileo, R.; Fusco, A.; Coppola, G.R.; et al. Genetics of Takotsubo Syndrome. Heart Fail. Clin. 2016, 12, 499–506. [Google Scholar] [CrossRef]
  92. de Gonzalo-Calvo, D.; Vea, A.; Bär, C.; Fiedler, J.; Couch, L.S.; Brotons, C.; Llorente-Cortes, V.; Thum, T. Circulating non-coding RNAs in biomarker-guided cardiovascular therapy: A novel tool for personalized medicine? Eur. Heart J. 2019, 40, 1643–1650. [Google Scholar] [CrossRef] [PubMed]
  93. d’Avenia, M.; Citro, R.; De Marco, M.; Veronese, A.; Rosati, A.; Visone, R.; Leptidis, S.; Philippen, L.; Vitale, G.; Cavallo, A.; et al. A novel miR-371a-5p-mediated pathway, leading to BAG3 upregulation in cardiomyocytes in response to epinephrine, is lost in Takotsubo cardiomyopathy. Cell Death Dis. 2015, 6, e1948. [Google Scholar] [CrossRef] [Green Version]
  94. Neil, C.; Nguyen, T.H.; Kucia, A.; Crouch, B.; Sverdlov, A.; Chirkov, Y.; Mahadavan, G.; Selvanayagam, J.; Dawson, D.; Beltrame, J.; et al. Slowly resolving global myocardial inflammation/oedema in Tako-Tsubo cardiomyopathy: Evidence from T2-weighted cardiac MRI. Heart 2012, 98, 1278–1284. [Google Scholar] [CrossRef] [PubMed]
  95. Nef, H.M.; Möllmann, H.; Troidl, C.; Kostin, S.; Böttger, T.; Voss, S.; Hilpert, P.; Krause, N.; Weber, M.; Rolf, A.; et al. Expression profiling of cardiac genes in Tako-Tsubo cardiomyopathy: Insight into a new cardiac entity. J. Mol. Cell. Cardiol. 2008, 44, 395–404. [Google Scholar] [CrossRef] [PubMed]
  96. Scally, C.; Rudd, A.; Mezincescu, A.; Wilson, H.; Srivanasan, J.; Horgan, G.; Broadhurst, P.; Newby, D.E.; Henning, A.; Dawson, D.K. Persistent Long-Term Structural, Functional, and Metabolic Changes After Stress-Induced (Takotsubo) Cardiomyopathy. Circulation 2018, 137, 1039–1048. [Google Scholar] [CrossRef] [PubMed]
  97. Surikow, S.Y.; Raman, B.; Licari, J.; Singh, K.; Nguyen, T.H.; Horowitz, J.D. Evidence of nitrosative stress within hearts of patients dying of Tako-tsubo cardiomyopathy. Int. J. Cardiol. 2015, 189, 112–114. [Google Scholar] [CrossRef] [PubMed]
  98. Nguyen, T.H.; Neil, C.J.; Sverdlov, A.L.; Ngo, D.T.; Chan, W.P.; Heresztyn, T.; Chirkov, Y.Y.; Tsikas, D.; Frenneaux, M.P.; Horowitz, J.D. Enhanced NO signaling in patients with takotsubo cardiomyopathy: Short-term pain, long-term gain? Cardiovasc. Drugs Ther. 2013, 27, 541–547. [Google Scholar] [CrossRef] [PubMed]
  99. Surikow, S.Y.; Nguyen, T.H.; Stafford, I.; Chapman, M.; Chacko, S.; Singh, K.; Licari, G.; Raman, B.; Kelly, D.J.; Zhang, Y.; et al. Nitrosative Stress as a Modulator of Inflammatory Change in a Model of Takotsubo Syndrome. JACC Basic Transl. Sci. 2018, 3, 213–226. [Google Scholar] [CrossRef] [PubMed]
  100. Cieślar-Pobuda, A.; Saenko, Y.; Rzeszowska-Wolny, J. PARP-1 inhibition induces a late increase in the level of reactive oxygen species in cells after ionizing radiation. Mutat. Res.-Fundam. Mol. Mech. Mutagen. 2012, 732, 9–15. [Google Scholar] [CrossRef]
  101. 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]
  102. Wilson, H.M.; Cheyne, L.; Brown, P.A.J.; Kerr, K.; Hannah, A.; Srinivasan, J.; Duniak, N.; Horgan, G.; Dawson, D.K. Characterization of the Myocardial Inflammatory Response in Acute Stress-Induced (Takotsubo) Cardiomyopathy. JACC Basic Transl. Sci. 2018, 3, 766–778. [Google Scholar] [CrossRef] [PubMed]
  103. Ederhy, S.; Dolladille, C.; Thuny, F.; Alexandre, J.; Cohen, A. Takotsubo syndrome in patients with cancer treated with immune checkpoint inhibitors: A new adverse cardiac complication. Eur. J. Heart Fail. 2019, 21, 945–947. [Google Scholar] [CrossRef]
  104. Lyon, A.R.; Yousaf, N.; Battisti, N.M.L.; Moslehi, J.; Larkin, J. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol. 2018, 19, e447–e458. [Google Scholar] [CrossRef]
  105. Kolodzinska, A.; Czarzasta, K.; Szczepankiewicz, B.; Glowczynska, R.; Fojt, A.; Ilczuk, T.; Budnik, M.; Krasuski, K.; Folta, M.; Cudnoch-Jedrzejewska, A.; et al. Toll-like receptor expression and apoptosis morphological patterns in female rat hearts with takotsubo syndrome induced by isoprenaline. Life Sci. 2018, 199, 112–121. [Google Scholar] [CrossRef] [PubMed]
  106. Rona, G. Catecholamine cardiotoxicity. J. Mol. Cell. Cardiol. 1985, 17, 291–306. [Google Scholar] [CrossRef]
  107. Szardien, S.; Mollmann, H.; Willmer, M.; Liebetrau, C.; Voss, S.; Troidl, C.; Hoffmann, J.; Rixe, J.; Elsasser, A.; Hamm, C.W.; et al. Molecular basis of disturbed extracellular matrix homeostasis in stress cardiomyopathy. Int. J. Cardiol. 2013, 168, 1685–1688. [Google Scholar] [CrossRef]
  108. Godsman, N.; Kohlhaas, M.; Nickel, A.; Cheyne, L.; Mingarelli, M.; Schweiger, L.; Hepburn, C.; Munts, C.; Welch, A.; Delibegovic, M.; et al. Metabolic alterations in a rat model of takotsubo syndrome. Cardiovasc. Res. 2022, 118, 1932–1946. [Google Scholar] [CrossRef] [PubMed]
  109. Oliveri, F.; Goud, H.K.; Mohammed, L.; Mehkari, Z.; Javed, M.; Althwanay, A.; Ahsan, F.; Rutkofsky, I.H. Role of Depression and Anxiety Disorders in Takotsubo Syndrome: The Psychiatric Side of Broken Heart. Cureus 2020, 12, e10400. [Google Scholar] [CrossRef] [PubMed]
  110. Katsuura, S.; Kuwano, Y.; Yamagishi, N.; Kurokawa, K.; Kajita, K.; Akaike, Y.; Nishida, K.; Masuda, K.; Tanahashi, T.; Rokutan, K. MicroRNAs miR-144/144* and miR-16 in peripheral blood are potential biomarkers for naturalistic stress in healthy Japanese medical students. Neurosci. Lett. 2012, 516, 79–84. [Google Scholar] [CrossRef]
  111. Bai, M.; Zhu, X.; Zhang, Y.; Zhang, S.; Zhang, L.; Xue, L.; Yi, J.; Yao, S.; Zhang, X. Abnormal Hippocampal BDNF and miR-16 Expression Is Associated with Depression-Like Behaviors Induced by Stress during Early Life. PLoS ONE 2012, 7, e46921. [Google Scholar] [CrossRef] [PubMed]
  112. Radu Enatescu, V.; Papava, I.; Enatescu, I.; Antonescu, M.; Anghel, A.; Seclaman, E.; Ovidiu Sirbu, I.; Marian, C. Circulating Plasma Miro RNAs in Patients with Major Deppresive Disorder Treated with Antidepressants: A Pilot Study. Psychiatry Investig. 2016, 13, 549–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. 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, 809–812. [Google Scholar] [CrossRef] [PubMed]
  114. 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]
  115. 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] [Green Version]
  116. 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, 1898–1908. [Google Scholar] [CrossRef] [PubMed]
  117. Pilgrim, T.M.; Wyss, T.R. Takotsubo cardiomyopathy or transient left ventricular apical ballooning syndrome: A systematic review. Int. J. Cardiol. 2008, 124, 283–292. [Google Scholar] [CrossRef] [PubMed]
  118. Bielecka-Dabrowa, A.; Mikhailidis, D.P.; Hannam, S.; Rysz, J.; Michalska, M.; Akashi, Y.J.; Banach, M. Takotsubo cardiomyopathy—The current state of knowledge. Int. J. Cardiol. 2010, 142, 120–125. [Google Scholar] [CrossRef] [PubMed]
  119. Inamasu, J.; Watanabe, E.; Okuda, K.; Kumai, T.; Sugimoto, K.; Ozaki, Y.; Hirose, Y. Are there differences between Takotsubo cardiomyopathy and neurogenic stunned myocardium? A prospective observational study. Int. J. Cardiol. 2014, 177, 1108–1110. [Google Scholar] [CrossRef] [PubMed]
  120. Guglin, M.; Novotorova, I. Neurogenic Stunned Myocardium and Takotsubo Cardiomyopathy Are the Same Syndrome: A Pooled Analysis. Congest. Hear. Fail. 2011, 17, 127–132. [Google Scholar] [CrossRef] [PubMed]
  121. Ancona, F.; Bertoldi, L.F.; Ruggieri, F.; Cerri, M.; Magnoni, M.; Beretta, L.; Cianflone, D.; Camici, P.G. Takotsubo cardiomyopathy and neurogenic stunned myocardium: Similar albeit different. Eur. Heart J. 2016, 37, 2830–2832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Biso, S.; Wongrakpanich, S.; Agrawal, A.; Yadlapati, S.; Kishlyansky, M.; Figueredo, V. A Review of Neurogenic Stunned Myocardium. Cardiovasc. Psychiatry Neurol. 2017, 2017, 5842182. [Google Scholar] [CrossRef] [Green Version]
  123. Madhavan, M.; Borlaug, B.A.; Lerman, A.; Rihal, C.S.; Prasad, A. Stress hormone and circulating biomarker profile of apical ballooning syndrome (Takotsubo cardiomyopathy): Insights into the clinical significance of B-type natriuretic peptide and troponin levels. Heart 2009, 95, 1436–1441. [Google Scholar] [CrossRef] [PubMed]
  124. Fröhlich, G.M.; Schoch, B.; Schmid, F.; Keller, P.; Sudano, I.; Lüscher, T.F.; Noll, G.; Ruschitzka, F.; Enseleit, F. Takotsubo cardiomyopathy has a unique cardiac biomarker profile: NT-proBNP/myoglobin and NT-proBNP/troponin T ratios for the differential diagnosis of acute coronary syndromes and stress induced cardiomyopathy. Int. J. Cardiol. 2012, 154, 328–332. [Google Scholar] [CrossRef] [PubMed]
  125. Omland, T.; Persson, A.; Ng, L.; O’Brien, R.; Karlsson, T.; Herlitz, J.; Hartford, M.; Caidahl, K. N-terminal pro-B-type natriuretic peptide and long-term mortality in acute coronary syndromes. Circulation 2002, 106, 2913–2918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Stiermaier, T.; Santoro, F.; Graf, T.; Guastafierro, F.; Tarantino, N. Prognostic value of N-Terminal Pro-B-Type Natriuretic Peptide in Takotsubo syndrome. Clin. Res. Cardiol. 2018, 107, 597–606. [Google Scholar] [CrossRef]
  127. Santoro, F.; Costantino, M.D.; Guastafierro, F.; Triggiani, G.; Ferraretti, A.; Tarantino, N.; Saguner, A.; Di Biase, M.; Brunetti, N.D. Inflammatory patterns in Takotsubo cardiomyopathy and acute coronary syndrome: A propensity score matched analysis. Atherosclerosis 2018, 274, 157–161. [Google Scholar] [CrossRef]
  128. Burgdorf, C.; Schubert, A.; Schunkert, H.; Kurowski, V.; Radke, P.W. Release patterns of copeptin and troponin in Tako-Tsubo cardiomyopathy. Peptides 2012, 34, 389–394. [Google Scholar] [CrossRef]
  129. Højagergaard, M.A.; Hassager, C.; Christensen, T.E.; Bang, L.E.; Gøtze, J.P.; Ostrowski, S.R.; Holmvang, L.; Frydland, M. Biomarkers in patients with Takotsubo cardiomyopathy compared to patients with acute anterior ST-elevation myocardial infarction. Biomarkers 2020, 25, 137–143. [Google Scholar] [CrossRef]
  130. Budnik, M.; Białek, S.; Peller, M.; Kiszkurno, A.; Kochanowski, J.; Kucharz, J.; Sitkiewicz, D.; Opolski, G. Serum copeptin and copeptin/NT-proBNP ratio—New tools to differentiate takotsubo syndrome from acute myocardial infarction. Folia Med. Cracov. 2020, 60, 5–14. [Google Scholar] [CrossRef]
  131. Nguyen, T.H.; Liu, S.; Ong, G.J.; Stafford, I.; Frenneaux, M.P.; Horowitz, J.D. Glycocalyx shedding is markedly increased during the acute phase of Takotsubo cardiomyopathy. Int. J. Cardiol. 2017, 243, 296–299. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Couch, L.S.; Channon, K.; Thum, T. Molecular Mechanisms of Takotsubo Syndrome. Int. J. Mol. Sci. 2022, 23, 12262. https://doi.org/10.3390/ijms232012262

AMA Style

Couch LS, Channon K, Thum T. Molecular Mechanisms of Takotsubo Syndrome. International Journal of Molecular Sciences. 2022; 23(20):12262. https://doi.org/10.3390/ijms232012262

Chicago/Turabian Style

Couch, Liam S., Keith Channon, and Thomas Thum. 2022. "Molecular Mechanisms of Takotsubo Syndrome" International Journal of Molecular Sciences 23, no. 20: 12262. https://doi.org/10.3390/ijms232012262

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