Next Article in Journal
In Vivo Comparison of Synthetic Macroporous Filamentous and Sponge-like Skin Substitute Matrices Reveals Morphometric Features of the Foreign Body Reaction According to 3D Biomaterial Designs
Previous Article in Journal
Depletion of VGLL4 Causes Perinatal Lethality without Affecting Myocardial Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Septic Cardiomyopathy: From Pathophysiology to the Clinical Setting

by
Federico Carbone
1,2,
Luca Liberale
1,2,
Alberto Preda
3,
Thomas Hellmut Schindler
4 and
Fabrizio Montecucco
1,2,*
1
First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
2
IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 16132 Genoa, Italy
3
Vita-Salute San Raffaele University, 20132 Milan, Italy
4
Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(18), 2833; https://doi.org/10.3390/cells11182833
Submission received: 27 July 2022 / Revised: 5 September 2022 / Accepted: 7 September 2022 / Published: 11 September 2022

Abstract

:
The onset of cardiomyopathy is a common feature in sepsis, with relevant effects on its pathophysiology and clinical care. Septic cardiomyopathy is characterized by reduced left ventricular (LV) contractility eventually associated with LV dilatation with or without right ventricle failure. Unfortunately, such a wide range of ultrasonographic findings does not reflect a deep comprehension of sepsis-induced cardiomyopathy, but rather a lack of consensus about its definition. Several echocardiographic parameters intrinsically depend on loading conditions (both preload and afterload) so that it may be challenging to discriminate which is primitive and which is induced by hemodynamic perturbances. Here, we explore the state of the art in sepsis-related cardiomyopathy. We focus on the shortcomings in its definition and point out how cardiac performance dynamically changes in response to different hemodynamic clusters. A special attention is also given to update the knowledge about molecular mechanisms leading to myocardial dysfunction and that recall those of myocardial hibernation. Ultimately, the aim of this review is to highlight the unsolved issue in the field of sepsis-induced cardiomyopathy as their implementation would lead to improve risk stratification and clinical care.

1. Introduction

Since 2016, sepsis has been defined as life-threatening organ dysfunction caused by a dysregulated host response to infection [1]. Such a paradigm shift emphasizes the supremacy of the non-homeostatic host response in infection pathophysiology. Septic patients may be now further categorized in different multidimensional phenotypes: at least four have been identified, not homologous with traditional patient clustering, but strongly correlated with patterns of host immune response and clinical outcomes [2]. When enduring, an inflammatory state may lead to persistent organ injury, and long-term mortality—even by several months to years—in elderly with highly comorbid conditions [3]. Meanwhile, a growing body of evidence is challenging the traditional view on septic cardiomyopathy. Historically assumed as adaptive response to intravascular hypovolemia, cardiac performance actually changes dynamically in accordance with different hemodynamic clusters [4]. Myocardial dysfunction is commonly found in sepsis and outlined by cellular abnormalities, circulating mediators and instrumental parameters. However, whether the degree of myocardial dysfunction is related to systemic organ failure and the extent to which sepsis-induced cardiomyopathy contributes to the outcome remains uncertain and somehow contradictory. This review article aims at summarizing the definition, epidemiology, pathophysiology, and diagnosis of septic cardiomyopathy. A special focus will be paid at identifying and addressing major clinical shortcomings.

2. Definition and Epidemiological Considerations

2.1. Challenges in Defining Septic Cardiomyopathy

Septic cardiomyopathy was first described in 1921—before the advent of antibiotics—by E. Romberg, in his Lehrbuch der Krankheiten des Herzens und der Blutgefäße (Textbook of Diseases of the Heart and Blood Vessels) as septische akute Myokarditis (septic acute myocarditis). It may be broadly defined as an acute cardiac dysfunction unrelated to ischemia that manifests in different ways: arrhythmias, left and/or right ventricular impairment during systole or diastole, with or without reduction in cardiac output [5,6,7] (Table 1).
Progress in cardiac imaging now makes available several parameters of cardiac performance—historically limited to ejection fraction assessment—but they have paradoxically increased the uncertainty on how to precisely define septic cardiomyopathy. Further issues challenging a global consensus on septic cardiomyopathy include: (i) the potential role of cardiovascular background as premorbid cardiac status; (ii) the need of assessing cardiac function in a setting of highly variable conditions, as determined by inotrope use, autonomic dysregulation of heart rate/arrhythmias, fluid challenge, rise in lactate and reduction in mixed venous oxygen saturation (Figure 1) [5,6,7]. Differentiating septic cardiomyopathy from Takotsubo cardiomyopathy is another relevant clinical issue. Although sepsis and septic shock may trigger Takotsubo cardiomyopathy, the underlying myocardial alterations are different. Furthermore, myocardial dysfunction in Takotsubo cardiomyopathy typically has a regional distribution, mainly an apical ballooning mimicking acute coronary syndrome. Yet, a right ventricular dysfunction cannot be ruled out. Furthermore, Takotsubo cardiomyopathy shows a normalization within days or weeks, although patients may suffer a worse prognosis [8,9,10,11,12,13,14]. The extent to which septic cardiomyopathy overlaps with myocarditis is also difficult to discriminate. Although they share histological markers of stress-induced cardiotoxicity (e.g., interstitial inflammatory infiltrate and edema in conjunction with myocytolysis, necrosis, and interstitial fibrosis), septic cardiomyopathy rarely presents with severe fulminant LV (left ventricular) dysfunction and is generally associated with full recovery of systolic performance [15]. Cardiac function during septic cardiomyopathy should be ultimately conceived as a dynamic assessment over time, not limited to echocardiographic parameters but rather considering the complex hemodynamic context. Gathering clinical, laboratory, and echocardiographic parameters, Geri and colleagues have recently identified five different cardiovascular phenotypes of septic cardiomyopathy (Table 1): (i) no evidence of any cardiac dysfunction; (ii) with LV systolic dysfunction; (iii) hyperkinetic profile, as defined by a preserved—or even supernormal LV systolic function—and an elevated aortic velocity time integral; (iv) right ventricle (RV) failure; (v) persistent hypovolemia [7]. Data-driven machine learning approaches are now unrevealing the importance of continuous monitoring cardiac function to define the correct hemodynamic phenotype [16]. Furthermore, the recent recognition of sepsis phenotypes may provide additional information—or even complicate—the definition and classification of sepsis cardiomyopathy [2]. The analysis of about thirty candidate variables across 1.3 million of patients from three large randomized clinical trials allowed identifying different patterns of organ dysfunction. Especially in the so-called δ phenotype—as compared with the α, β, and γ ones—the cardiovascular involvement was prevalent and associated with increase in inflammatory markers and in abnormal coagulation. Not surprisingly, this phenotype with prevalent cardiovascular involvement was characterized by significantly higher mortality rates. Although δ phenotype is well represented by the classic SOFA score, its characterization deserves more investigations that would also improve definition, characterization, pathophysiology and, hopefully, risk of sepsis cardiomyopathy.

2.2. Epidemiology of Septic Cardiomyopathy

Studies on septic cardiomyopathy report a broad range of prevalence ranging from 10% to 70%. Lacking a broad consensus on definition, the epidemiology of septic cardiomyopathy remains elusive. Such a variability reflects the definition used in study designs and the burden of confounding factors: male sex, age, high lactate levels at admission and pre-existing cardiac performance [5]. Long-term effects of septic cardiomyopathy are also poorly addressed but the prevalence of mortality seems to be higher [17] and characterized by a U-shaped non-linear distribution [18]. The timeline raises further issues. The relatively few studies published before and after the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) are potentially biased in patient selection [17,19,20]. Most of the them are limited to the setting of intensive care unit (ICU), where patients are somehow selected and largely differ from those admitted in internal medicine wards in terms of comorbidities, standards of care and outcome. Cumulative rate of sepsis admitted to the ICU is indeed quite low (44 vs. 367 cases per 100,000 adult/year) [21] and patients are generally younger and with better baseline myocardial performance. Of note, the risk of cardiomyopathy seems to be higher in ICU, as older comorbid patients admitted to internal medicine wards are likely to die before developing cardiac dysfunction. Based on the above considerations, a consensus definition of septic cardiomyopathy is claimed as a milestone for future studies.

3. Pathophysiology of Septic Cardiomyopathy

Endothelial, metabolic, and immune response abnormalities are generally involved in the pathogenesis of ventricular dysfunction and arrhythmias during sepsis, whereas the potential role of myocardial ischemia seems limited. Impaired blood flow autoregulation in coronary microcirculation and altered metabolism of lactate, free fatty acid, and glucose likely play a leading role. Septic myocytes may indeed switch their primary energy substrate from free fatty acids to glucose with detrimental effects on contractility, similarly to that observed during post-ischemic myocardial hibernation [22]. However, the transition point from a protective to an adverse and maladaptive process is missing the humoral response in sepsis (Figure 2).

3.1. Inflammatory Pathways and Cardiomyocyte Dysfunction

The hyperinflammatory—not counterbalanced—response is historically reported as a paradigm of sepsis. Early clinical trials then focused on blunting inflammatory response by common anti-inflammatory drugs such as glucocorticoids, non-steroidal anti-inflammatory drugs or target therapy, with very limited results [23]. The extreme variability in pro-/anti-inflammatory balance and the wide number of molecular pathways may explain such disheartening results [24,25]. Ancestral signals released by pathogens (pathogen-associated molecular patterns, PAMPs) or by damaged host tissues (damage-associated molecular patterns, DAMPs) and receptors such as Toll-like receptors (TLRs) trigger multiple intracellular pathways, including the activation of nuclear factor-kB (NF-kB) and mitogen-activated protein kinase (MAPK) [26,27,28]. The DAMPs with a recognized role in the pathophysiology of septic cardiomyopathy include: heparan sulphate, which increases intranuclear transcription of pro-inflammatory cytokines and vascular permeability [29]; high mobility group protein B1, which induces loss of calcium from sarcoplasmic reticulum [30]; and histones, which are able to interfere with the production of cellular ATP by reducing mitochondrial membrane potential and causing LV dysfunction and arrhythmias [31]. Among PAMPs, bacterial lipopolysaccharide (LPS) partially mimics hemodynamic effects of septic shock when administered to animal models or humans [32,33]. Detrimental effects of endotoxemia on cardiac contractility are largely due to mitochondrial dysfunction that leads to abnormal calcium handling, disruption of ATP synthesis, endothelium reticulum stress, and autophagy. Furthermore, LPS leads to electrophysiological dysfunctions through a both a direct and cytokine-mediated effect on sodium current kinetics and non-selective cation channel transient receptor potential vanilloid 1 (TRPV1) [34,35,36,37]. Furthermore, LPS and its downstream mediator TNF-α, IL-1β, IL-6, and C5a and ROS [38,39] critically alter calcium (Ca++) homeostasis by blunting the amplitude of intracellular currents and concentrations. Dysfunction of intracellular calcium transporters and decreased calcium sensitivity in cardiac myofilaments (due to phosphorylation of inhibitory troponin I) are cornerstones of the impaired excitation-contraction coupling. Ultimately, endotoxemia impairs sarcolemma diastolic Ca++ extrusion with consequent overload [40,41,42,43] that determines systolic and diastolic dysfunction during sepsis [44,45]. The characteristic delayed reversibility of septic cardiomyopathy might be explained by the synthesis of new myofilaments to replace the previously phosphorylated (and therefore inactive) ones.
Signals regulated by the TLRs expressed on myocyte surface (TLR2, TRL3, TLR4 and TLR9) lead to transcription of several pro-inflammatory cardiac depressive factors and cytokines such as interleukin (IL)-1, IL-6, tumor necrosis factor (TNF)-α and complement anaphylatoxin C5a [46,47]. LV ejection fraction (LVEF) was found higher in septic mouse with TRL3 and TLR9 gene deletion, suggesting their role in the mechanism of sepsis-induced cardiac dysfunction [48]. Likewise, TLR4 regulates oxidative stress in ryanodine receptor 2 involved in the storage of calcium within sarcoplasmic reticulum of cardiomyocytes [49], and its inhibition showed protective effects [50]. C5a is so far the only complement factor with reported direct myocardial depressive effect, despite several cardiac enzymes related to cardiac dysfunction during sepsis, such as serca2, NCX and Na+/K+-atpase, which are complement receptor dependent [51]. Finally, adhesion molecules take part in homing mechanisms and are upregulated during inflammatory response. In murine coronary endothelium and cardiomyocytes, specific adhesion molecules were upregulated after LPS and TNF-α infusion. Moreover, the use of antibody blocking these molecules such as anti-Intercellular adhesion molecule-1 (ICAM-1) and vascular adhesion molecule-1 (VCAM-1) showed to prevent myocardial neutrophil accumulation and cardiac dysfunction in animal models of sepsis [52]. On the contrary, neutrophils depletion did not protect against myocardial dysfunction during sepsis, suggesting their lesser cardiotoxic potential.
Nitric oxide (NO) is synthesized by nitric oxide synthase (NOS) in different cells of the CV system including cardiac myocytes, and it exerts important roles in maintaining tissue homeostasis by reducing oxidative stress. Indeed, via the cGMP pathway, NO regulates vascular tone, has an antioxidant effect, inhibits leukocyte and platelet adhesion to the endothelium, and increases myocardial contractility [53]. Inflammation and oxidative stress show deep interplay, fueling each other and facilitating the onset of deleterious vicious circles in different diseases including sepsis. Differently from its endothelial and neuronal isoforms, inducible NOS (iNOS) is not constitutively active and when highly expressed is a major responsible for vasodilatation and hypotension in shock [54]. Indeed, iNOS can produce large amount of NO when an inflammatory response occurs [55]. Sepsis leads to overexpression of iNOS not only in immune cells but also in the myocardium [56]. Such increased expression has detrimental effects on contractile function of cardiomyocytes, in part through the paradoxical induction of reactive oxygen species (ROS), i.e., peroxynitrite [57], in part through the down-regulation of adrenaline receptors and decreased sensitivity to calcium [29]. Confirming the important role played by iNOS in myocardial dysfunction during sepsis, several non-specific anti-NOS drugs such as melatonin and methylene blue showed beneficial effects in terms of cardiovascular function and prognosis in preclinical studies [57,58].

3.2. Adrenergic System

Increased activation of the sympathetic system is a major compensatory reaction to septic-related vasoplegia. During sepsis, β-adrenergic receptors (βARs) are downregulated, and the responsiveness to catecholamines is reduced in the whole cardiovascular system [59,60]. When persistent, this adaptive response become maladaptive [61]. In myocardial tissue, excessive stimulation of cardiac βARs suppresses their expression and leads to an inhibitory response with reverse of adrenergic G protein coupling [62], intracellular calcium overload [63], increased production of ROS [64], disruption of membrane potential via the inhibition of Na+/K+-ATPase pump [65], and induction of apoptosis [66]. A relevant role is played by Cardiac G Protein-Coupled Receptor Kinase 2 (GRK2). They are the major negative regulators of βAR pro-contractile signal in sepsis-induced myocardial dysfunction [67] and other different cardiac diseases including chronic heart failure and Takotsubo syndrome [68].

3.3. Microvascular Dysfunction and Vasoactive Peptides

Activation of endothelial cells in consequence of an infective stimulus leads to the synthesis of inflammatory cytokines, increased expression of cell adhesion molecules, loss of the barrier function by glycocalyx shedding, edema, apoptosis, hypercoagulative state and vasoplegia [69]. Several endogenous vasoactive peptides generate and maintain endothelial dysregulation. LPS and cytokines up-regulate cyclooxygenase (COX)-2 (inducible form) and then the production of prostanoids from arachidonic acid. This process mainly occurs in inflammatory cells, myofibroblasts, endothelium and even in cardiomyocytes (i.e., in myocardial infarction) [70,71]. Elevated levels of prostanoids such as thromboxane and prostacyclin are associated with coronary microvascular dysfunction [72]. However, clinical trials have failed so far in proving any beneficial effect of pharmacological COX inhibition on coronary microvascular homeostasis in sepsis. Less is known about the potential role of endothelin-1 (ET-1) in sepsis cardiomyopathy, whereas an increase has been reported in chronic cardiac disease (i.e., heart failure) [73]. ET-1 would have a major role in infectious disease, as a promoter of cytokines release, platelet aggregation and vasoreactivity [74]. In single mouse study, cardiac overexpression of ET-1 was associated with the onset of severe sepsis cardiomyopathy, characterized by interstitial infiltration of macrophages and T lymphocytes and increased levels of the pro-inflammatory cytokines TNF-α, INF-γ, IL-1, and IL-6 [75].

3.4. Energetic Dysmetabolism

Sepsis is characterized by altered myocardial lipoprotein metabolism and mobilization of triglycerides and free fatty acids to overwhelm the systemic suppression of energy production [76]. Physiologically, about 70% of cardiac ATP is produced via lipid oxidation, while the rest is produced via glucose oxidation. A minor part also derives from the catabolism of lactate and ketone bodies. Under pathological conditions, glucose oxidation becomes the prevalent energetic pathway [77]. During sepsis, a reduction in fatty acid oxidation is not compensated by the increase in glucose catabolism due to altered insulin action, exacerbation of the inhibitory effects of alternative substrates on glycolysis, and blunted glycogen synthesis [78]. Several enzymes involved in intracellular cardiac fatty acid mobilization and oxidation are also inhibited [79,80]. Cardiac transcriptional factors associated with fatty acid oxidation, such as peroxisome proliferator activated receptors (PPARs) are also suppressed in sepsis and their stimulation with PPARs agonists lead to improved survival in septic mice [81]. Mitochondrial dysfunction further contributes to the energetic failure in sepsis cardiomyopathy, thus being a non-negligible cause of reduced outcome [82,83,84]. Inflammation and oxidative stress alter mitochondrial structure determining swelling, cytoplasmic accumulation of denatured protein and lysosomal lesions [85,86]. Such a damage interferes with the respiratory chain [87] with falling in ATP synthesis, release of calcium and pro-apoptotic proteins [88]. The pro-oxidant environment induced by endotoxemia also trigger cardiac mitophagy, a defensive mechanism for the removal of damaged mitochondria [89]. The antioxidant N-acetylcysteine seems to have a protective roles against contractile dysfunction and mitophagy. Even levosimendam in both mice and human beings has beneficial effects due to calcium sensitization and antioxidant properties [90]. Lastly, when the degree of mitochondrial dysfunction is mild, myocardial hibernation may occur. This is an adaptive, self-protective mechanism during which all functions get reduced. Such a downregulation of mitochondrial gene transcription has been reported in sepsis cardiomyopathy [91] and represents a reversible condition that improves together with the resolution of sepsis [84].

4. Clinical Diagnosis

A combination of clinical, biochemical and hemodynamic parameters is increasingly described as the best approach to identify and stratify septic cardiomyopathy.

4.1. Doppler Echocardiography: From Classic to Innovative Assessment

Echocardiography is considered the first line approach for the global assessment of patients with septic shock. The use of ultrasound in this field has progressed over years but the application of echocardiography for diagnosis/monitoring of septic cardiomyopathy remains challenging. The dependence on loading conditions is a not overcoming intrinsic limitation for several echocardiographic parameters, which rather reflect the degree of fluid resuscitation/vascular leak (i.e., preload) and vasoplegia/response to vasoactive drugs (i.e., afterload) [7]. This is especially true for LVEF, traditionally considered the hallmark of septic cardiomyopathy. Indeed, merely identifying low LVEF (<50%) during sepsis does not have predictive value in these patients [92]. Rather, LVEF would reflect the abovementioned U-shaped non-linear distribution. Indeed even a hyperdynamic LVEF—mostly expression of reduced afterload—is burdened by higher short-term mortality [93]. As partially afterload-independent measures, tissue Doppler imaging (TDI) and speckle-tracking echocardiography (STE) are promising candidates. Unlike the peak systolic velocity of the mitral annulus (referred to as S’) [20], the global longitudinal strain (GLS) is substantially more performing. GLS is a semi-automated algorithm tracking selected region of myocardium during cardiac cycle (more specifically the ratio of the maximal change in the myocardial longitudinal length in systole to the original length in diastole). GLS may detect the impairment of LV systolic function before LVEF declines and worse GLS values are associated with higher mortality in patients with severe sepsis and septic shock [94]. Looking at the future, coupling afterload and cardiac performance might optimize echocardiographic assessment by overcoming this critical bias.
A rise in end-diastolic volume is common in septic shock patients—especially in those receiving large volemic replacement—and likely associated with poor outcome. The rationale for considering diastolic dysfunction in sepsis-induced cardiomyopathy relies on the upstream effect on pulmonary circulation, right heart, and finally tissue edema. However, the mining of diastolic dysfunction in sepsis cardiomyopathy remains to be elucidated. The concept of sepsis-induced cardiomyopathy is originally based on the abnormal LV function, but the risk ratio of patients with concomitant diastolic dysfunction is not yet established. In particular, the TDI-derived velocity of the mitral annulus during early diastole (e’)—considered relatively preload independent—and the ratio with the peak of trans-mitral inflow velocity (E) are substantially associated with mortality [95]. However, pathophysiological and therapeutic implications of diastolic dysfunction are still weak. The assessment of diastolic function is not yet validated for critically ill patients, and data extrapolation is very challenging when the increase in LV end-diastolic pressure is influenced by underlying cardiac performance (frequently unknown) and a variable degree of fluid resuscitation/vascular leak). Furthermore, mitral annulus velocity is influenced by heart rate in hyperdynamic cardiac output.
Right ventricle (RV) dysfunction is increasingly being reported in patients with sepsis. It can be defined as a fractional area change (FAC) less than 35% or tricuspid annulus systolic plane excursion (TAPSE)—the most reproducible measure—less than 16 mm. Systolic RV pressure may also be easily calculated from tricuspid regurgitation flow. RV dysfunction is common in severe sepsis/septic shock. Its prevalence ranges from 40% to 60% at admission but significantly increase when the enrollment lasts up to 72 h. RV dysfunction correlated with longer stay in ICU, and independently associated with higher risk of both early and long-term mortality [96,97,98]. Nevertheless, conflicting results exist and the complex geometry of RV—alongside the tight ventricular interdependence—raises many questions. RV is a thin-walled structure facing off a high-compliance/low-resistance pulmonary circulation. In sepsis, a paradoxical increase in pulmonary vasculature resistance may occur, due to hypoxic vasoconstriction, pulmonary source of infection/acute respiratory distress syndrome (ARDS), hypercapnia, and mechanical ventilation. Fluid overload further increases the preload on RV, leading to isolated RV dysfunction. In addition, LV dysfunction remains the leading cause of RV dysfunction and the association with poor outcome might be rather a “false positive” [99].

4.2. Cardiac Performance

The afterload-related cardiac performance (ACP) is a relative old concept firstly introduced in 2011 and defined as [100]:
ACP (%) = COmeasured/COpredicted × 100%
In turn, Cardiac Output (CO)predicted may be calculated as a function of systemic vascular resistance (SVR):
COpredicted = β0 × SVRβ1 (β0 = 394.07, β1 = −0.64)
In preliminary studies, ACP estimation correlates with both APACHE II and SOFA scores, even improving mortality prediction in both early and late stages of sepsis. ACP is now considered as valid prognostic marker in sepsis and septic shock, ACP values lower than 80% are associated with a higher risk of mortality in septic patients [101,102]. When defining septic cardiomyopathy by ACP (<80%), a recent study from Wilhelm and coll. reported that about 50% of patients with septic shock have at least a moderate septic cardiomyopathy, and that the lower the ACP the higher is mortality in this group of patients [103]. While validation in larger studies is needed, the use of a machine learning approach to implement this algorithm might further improve the model’s performance. Diameter and collapsibility of inferior vena cava and B line quantification at lung ultrasound are further intriguing surrogate measures of cardiac performance that deserve future investigations.

4.3. Biomarkers

The lack of a consensus definition and the uncertainties on pathophysiology of septic cardiomyopathy makes it difficult to identify and validate biomarkers of disease. Being associated with cardiac injury, troponins and natriuretic peptides rise during sepsis and correlate with the degree of myocardial dysfunction [104]. The mechanisms of troponin release during sepsis remain to be fully elucidated, but inflammation, myocardial wall stress, drug toxicity and kidney dysfunction rather than cell death are likely be implicated. Newly developed echocardiographic parameters, including TDI e STE applied to both left and right ventricles correlated with troponin levels [105], but their association with mortality likely reflects the severity of disease rather than the effective degree of myocardial dysfunction [94].
Brain natriuretic peptides (BNP and NT-proBNP) are secreted by myocardial cells in response to stretch. This would candidate BNPs as surrogate biomarkers of LV dysfunction. A significant association with both systolic and diastolic LV dysfunction was actually—but not unequivocally—demonstrated for BNPs. Preliminary data also link the rise in BNPs with RV dysfunction [106]. However, increased BNP levels during sepsis may be due to different conditions including lung involvement (e.g., source of infection, ARDS, mechanical ventilation), pre-existing cardiac dysfunction, and renal impairment. BNPs also failed to predict the responsiveness to fluid challenge, and cannot be used as a reliable biomarker of volemic status during severe sepsis/septic shock 98. Even concerning mortality, the rise in BNPs is associated with higher risk in meta-analyses but with substantial heterogeneity across the studies [107,108]. As for acute decompensated heart failure, serial measurement of BNPs might improve the performance of these biomarkers and clarify whether the prognostic value is specifically tied with cardiac dysfunction or rather with sepsis severity.
The emerging field of non-coding RNAs (ncRNAs) recently opened new scenarios for modulation of inflammation in sepsis, including sepsis-related cardiomyopathy 101. Among the wide array of ncRNA, a massive amount of experimental data concerns microRNAs (miRNAs) with good potential for clinical translation [109]. Among the growing number of miRNAs with clinical interest for sepsis/induced cardiomyopathy, the best predictors of cardiac injury are miR21-3p and miR-155, but correlations with myocardial dysfunction degree are also reported for miR-135a and miR-494-3p.
The cytokine storm makes it difficult to identify biomarkers of sepsis cardiomyopathy across cytokines/interleukins. A single observational study has reported an association with IL-8 [110]. Something more might be expected from artificial intelligence in the next future. Bioinformatics analyses has the potential for deciphering this complex signals. Tumor necrosis factor, Jak-signal transducer and activator of transcription (STAT), hypoxia-inducible transcription factor-1, and their interactions are increasingly described as crossroads of sepsis cardiomyopathy [111].

5. Conclusions and Outlook

The most important goal in the field of septic cardiomyopathy might be its recognition. The lack of a consensus definition led to divergence about its clinical relevance, whereas the advance in echocardiographic technology/algorithm is worryingly—and paradoxically—tangling up the search for a general agreement. Of paramount importance for standardizing the definition of septic cardiomyopathy would be to discriminate whether this is primitive and how it can induce hemodynamic perturbances. In addition, the dynamicity of changes in myocardial function should be considered. There is also growing awareness that mild declines in cardiac performance are rather to be considered as an adaptive response. In line with these missing points, studies outlining treatment goals for sepsis cardiomyopathy are lacking. The “early goal-directed therapy” still remains the current approach, since 2001, although this was not confirmed in later clinical trials (e.g., ProCESS, ARISE, ProMISe) [112,113,114]. As for sepsis, infection control and optimization of hemodynamic parameters are also considered the standard treatment for sepsis cardiomyopathy. Even the assumption of complete recovery in the long-term after the sepsis need to be revised. Long-term outcomes need to be added in study protocols, and this requires adequate infrastructures for monitoring cardiovascular health status after severe sepsis/septic shock. Overall, those unsolved questions have so far limited any therapeutic implications. Implementing those unsolved issues might improve risk stratification and ultimately promote the translation of therapeutic approaches now confined to the experimental field.

Author Contributions

All authors contributed by writing (F.C. and A.P.), reviewing (T.H.S. and F.M.) and editing (L.L.) this review article. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the Rete Cardiologica of Italian Ministry of Health (#2754291) to Federico Carbone. The research was funded by a grant from the Internal Medicine Department of the University of Genoa to Federico Carbone.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef] [PubMed]
  2. Seymour, C.W.; Kennedy, J.N.; Wang, S.; Chang, C.H.; Elliott, C.F.; Xu, Z.; Berry, S.; Clermont, G.; Cooper, G.; Gomez, H.; et al. Derivation, validation, and potential treatment implications of novel clinical phenotypes for sepsis. JAMA 2019, 321, 2003–2017. [Google Scholar] [CrossRef] [PubMed]
  3. Delano, M.J.; Ward, P.A. Sepsis-induced immune dysfunction: Can immune therapies reduce mortality? J. Clin. Investig. 2016, 126, 23–31. [Google Scholar] [CrossRef]
  4. Daulasim, A.; Vieillard-Baron, A.; Geri, G. Hemodynamic clinical phenotyping in septic shock. Curr. Opin. Crit. Care 2021, 27, 290–297. [Google Scholar] [CrossRef] [PubMed]
  5. Beesley, S.J.; Weber, G.; Sarge, T.; Nikravan, S.; Grissom, C.K.; Lanspa, M.J.; Shahul, S.; Brown, S.M. Septic cardiomyopathy. Crit. Care Med. 2018, 46, 625–634. [Google Scholar] [CrossRef]
  6. Martin, L.; Derwall, M.; Al Zoubi, S.; Zechendorf, E.; Reuter, D.A.; Thiemermann, C.; Schuerholz, T. The septic heart: Current understanding of molecular mechanisms and clinical implications. Chest 2019, 155, 427–437. [Google Scholar] [CrossRef]
  7. Geri, G.; Vignon, P.; Aubry, A.; Fedou, A.L.; Charron, C.; Silva, S.; Repesse, X.; Vieillard-Baron, A. Cardiovascular clusters in septic shock combining clinical and echocardiographic parameters: A post hoc analysis. Intensive Care Med. 2019, 45, 657–667. [Google Scholar] [CrossRef]
  8. El-Battrawy, I.; Lang, S.; Ansari, U.; Tulumen, E.; Schramm, K.; Fastner, C.; Zhou, X.; Hoffmann, U.; Borggrefe, M.; Akin, I. Prevalence of malignant arrhythmia and sudden cardiac death in takotsubo syndrome and its management. Europace 2018, 20, 843–850. [Google Scholar] [CrossRef]
  9. Vallabhajosyula, S.; Deshmukh, A.J.; Kashani, K.; Prasad, A.; Sakhuja, A. Tako-tsubo cardiomyopathy in severe sepsis: Nationwide trends, predictors, and outcomes. J. Am. Heart Assoc. 2018, 7, e009160. [Google Scholar] [CrossRef]
  10. Dias, A.; Nunez Gil, I.J.; Santoro, F.; Madias, J.E.; Pelliccia, F.; Brunetti, N.D.; Salmoirago-Blotcher, E.; Sharkey, S.W.; Eitel, I.; Akashi, Y.J.; et al. Takotsubo syndrome: State-of-the-art review by an expert panel—Part 1. Cardiovasc. Revasc. Med. 2019, 20, 70–79. [Google Scholar] [CrossRef]
  11. Dias, A.; Nunez Gil, I.J.; Santoro, F.; Madias, J.E.; Pelliccia, F.; Brunetti, N.D.; Salmoirago-Blotcher, E.; Sharkey, S.W.; Eitel, I.; Akashi, Y.J.; et al. Takotsubo syndrome: State-of-the-art review by an expert panel—Part 2. Cardiovasc. Revasc. Med. 2019, 20, 153–166. [Google Scholar] [CrossRef] [PubMed]
  12. El-Battrawy, I.; Santoro, F.; Stiermaier, T.; Moller, C.; Guastafierro, F.; Novo, G.; Novo, S.; Santangelo, A.; Mariano, E.; Romeo, F.; et al. Prevalence, management, and outcome of adverse rhythm disorders in takotsubo syndrome: Insights from the international multicenter geist registry. Heart Fail. Rev. 2020, 25, 505–511. [Google Scholar] [CrossRef] [PubMed]
  13. El-Battrawy, I.; Santoro, F.; Stiermaier, T.; Moller, C.; Guastafierro, F.; Novo, G.; Novo, S.; Mariano, E.; Romeo, F.; Romeo, F.; et al. Incidence and clinical impact of right ventricular involvement (biventricular ballooning) in takotsubo syndrome: Results from the geist registry. Chest 2021, 160, 1433–1441. [Google Scholar] [CrossRef]
  14. El-Battrawy, I.; Cammann, V.L.; Kato, K.; Szawan, K.A.; Di Vece, D.; Rossi, A.; Wischnewsky, M.; Hermes-Laufer, J.; Gili, S.; Citro, R.; et al. Impact of atrial fibrillation on outcome in takotsubo syndrome: Data from the international takotsubo registry. J. Am. Heart Assoc. 2021, 10, e014059. [Google Scholar] [CrossRef] [PubMed]
  15. Schmittinger, C.A.; Dunser, M.W.; Torgersen, C.; Luckner, G.; Lorenz, I.; Schmid, S.; Joannidis, M.; Moser, P.; Hasibeder, W.R.; Halabi, M.; et al. Histologic pathologies of the myocardium in septic shock: A prospective observational study. Shock 2013, 39, 329–335. [Google Scholar] [CrossRef]
  16. Liu, R.; Greenstein, J.L.; Granite, S.J.; Fackler, J.C.; Bembea, M.M.; Sarma, S.V.; Winslow, R.L. Data-driven discovery of a novel sepsis pre-shock state predicts impending septic shock in the icu. Sci. Rep. 2019, 9, 6145. [Google Scholar] [CrossRef] [PubMed]
  17. Vallabhajosyula, S.; Shankar, A.; Vojjini, R.; Cheungpasitporn, W.; Sundaragiri, P.R.; DuBrock, H.M.; Sekiguchi, H.; Frantz, R.P.; Cajigas, H.R.; Kane, G.C.; et al. Impact of right ventricular dysfunction on short-term and long-term mortality in sepsis: A meta-analysis of 1,373 patients. Chest 2021, 159, 2254–2263. [Google Scholar] [CrossRef]
  18. Beesley, S.J.; Sorensen, J.; Walkey, A.J.; Tonna, J.E.; Lanspa, M.J.; Hirshberg, E.; Grissom, C.K.; Horne, B.D.; Burk, R.; Abraham, T.P.; et al. Long-term implications of abnormal left ventricular strain during sepsis. Crit. Care Med. 2021, 49, e444–e453. [Google Scholar] [CrossRef] [PubMed]
  19. Huang, S.J.; Nalos, M.; McLean, A.S. Is early ventricular dysfunction or dilatation associated with lower mortality rate in adult severe sepsis and septic shock? A meta-analysis. Crit. Care 2013, 17, R96. [Google Scholar] [CrossRef]
  20. Sanfilippo, F.; Huang, S.; Messina, A.; Franchi, F.; Oliveri, F.; Vieillard-Baron, A.; Cecconi, M.; Astuto, M. Systolic dysfunction as evaluated by tissue doppler imaging echocardiography and mortality in septic patients: A systematic review and meta-analysis. J. Crit. Care 2021, 62, 256–264. [Google Scholar] [CrossRef]
  21. Esteban, A.; Frutos-Vivar, F.; Ferguson, N.D.; Penuelas, O.; Lorente, J.A.; Gordo, F.; Honrubia, T.; Algora, A.; Bustos, A.; Garcia, G.; et al. Sepsis incidence and outcome: Contrasting the intensive care unit with the hospital ward. Crit. Care Med. 2007, 35, 1284–1289. [Google Scholar] [CrossRef] [PubMed]
  22. Levy, R.J.; Piel, D.A.; Acton, P.D.; Zhou, R.; Ferrari, V.A.; Karp, J.S.; Deutschman, C.S. Evidence of myocardial hibernation in the septic heart. Crit. Care Med. 2005, 33, 2752–2756. [Google Scholar] [CrossRef] [PubMed]
  23. Cavaillon, J.M.; Singer, M.; Skirecki, T. Sepsis therapies: Learning from 30 years of failure of translational research to propose new leads. EMBO Mol. Med. 2020, 12, e10128. [Google Scholar] [CrossRef] [PubMed]
  24. Venet, F.; Davin, F.; Guignant, C.; Larue, A.; Cazalis, M.A.; Darbon, R.; Allombert, C.; Mougin, B.; Malcus, C.; Poitevin-Later, F.; et al. Early assessment of leukocyte alterations at diagnosis of septic shock. Shock 2010, 34, 358–363. [Google Scholar] [CrossRef] [PubMed]
  25. Davies, R.; O’Dea, K.; Gordon, A. Immune therapy in sepsis: Are we ready to try again? J. Intensive Care Soc. 2018, 19, 326–344. [Google Scholar] [CrossRef] [PubMed]
  26. Conway-Morris, A.; Wilson, J.; Shankar-Hari, M. Immune activation in sepsis. Crit. Care Clin. 2018, 34, 29–42. [Google Scholar] [CrossRef] [PubMed]
  27. Hawiger, J. Heartfelt sepsis: Microvascular injury due to genomic storm. Kardiol. Pol. 2018, 76, 1203–1216. [Google Scholar] [CrossRef]
  28. Rosengart, M.R.; Nathens, A.B.; Arbabi, S.; Neff, M.J.; Garcia, I.; Martin, T.R.; Maier, R.V. Mitogen-activated protein kinases in the intensive care unit: Prognostic potential. Ann. Surg. 2003, 237, 94–100. [Google Scholar] [CrossRef]
  29. Martin, L.; Schmitz, S.; De Santis, R.; Doemming, S.; Haase, H.; Hoeger, J.; Heinbockel, L.; Brandenburg, K.; Marx, G.; Schuerholz, T. Peptide 19-2.5 inhibits heparan sulfate-triggered inflammation in murine cardiomyocytes stimulated with human sepsis serum. PLoS ONE 2015, 10, e0127584. [Google Scholar] [CrossRef]
  30. Wang, H.; Ward, M.F.; Sama, A.E. Novel hmgb1-inhibiting therapeutic agents for experimental sepsis. Shock 2009, 32, 348–357. [Google Scholar] [CrossRef] [Green Version]
  31. Alhamdi, Y.; Abrams, S.T.; Cheng, Z.; Jing, S.; Su, D.; Liu, Z.; Lane, S.; Welters, I.; Wang, G.; Toh, C.H. Circulating histones are major mediators of cardiac injury in patients with sepsis. Crit. Care Med. 2015, 43, 2094–2103. [Google Scholar] [CrossRef] [PubMed]
  32. Suffredini, A.F.; Fromm, R.E.; Parker, M.M.; Brenner, M.; Kovacs, J.A.; Wesley, R.A.; Parrillo, J.E. The cardiovascular response of normal humans to the administration of endotoxin. N. Engl. J. Med. 1989, 321, 280–287. [Google Scholar] [CrossRef] [PubMed]
  33. Danner, R.L.; Elin, R.J.; Hosseini, J.M.; Wesley, R.A.; Reilly, J.M.; Parillo, J.E. Endotoxemia in human septic shock. Chest 1991, 99, 169–175. [Google Scholar] [CrossRef]
  34. Yucel, G.; Zhao, Z.; El-Battrawy, I.; Lan, H.; Lang, S.; Li, X.; Buljubasic, F.; Zimmermann, W.H.; Cyganek, L.; Utikal, J.; et al. Lipopolysaccharides induced inflammatory responses and electrophysiological dysfunctions in human-induced pluripotent stem cell derived cardiomyocytes. Sci. Rep. 2017, 7, 2935. [Google Scholar] [CrossRef]
  35. Sattler, K.; El-Battrawy, I.; Cyganek, L.; Lang, S.; Lan, H.; Li, X.; Zhao, Z.; Utikal, J.; Wieland, T.; Borggrefe, M.; et al. Trpv1 activation and internalization is part of the lps-induced inflammation in human ipsc-derived cardiomyocytes. Sci. Rep. 2021, 11, 14689. [Google Scholar] [CrossRef] [PubMed]
  36. Liao, Z.; Li, Y.; Fan, X.; Yang, Z.; El-Battrawy, I.; Zhou, X.; Akin, I. Lipopolysaccharide modifies sodium current kinetics through ros and pkc signalling in induced pluripotent stem-derived cardiomyocytes from brugada syndrome patient. J. Cardiovasc. Dev. Dis. 2022, 9, 119. [Google Scholar] [CrossRef]
  37. Fan, X.; Yang, G.; Kowitz, J.; Akin, I.; Zhou, X.; El-Battrawy, I. Takotsubo syndrome: Translational implications and pathomechanisms. Int. J. Mol. Sci. 2022, 23, 1951. [Google Scholar] [CrossRef] [PubMed]
  38. Natanson, C.; Eichenholz, P.W.; Danner, R.L.; Eichacker, P.Q.; Hoffman, W.D.; Kuo, G.C.; Banks, S.M.; MacVittie, T.J.; Parrillo, J.E. Endotoxin and tumor necrosis factor challenges in dogs simulate the cardiovascular profile of human septic shock. J. Exp. Med. 1989, 169, 823–832. [Google Scholar] [CrossRef]
  39. Kalbitz, M.; Fattahi, F.; Grailer, J.J.; Jajou, L.; Malan, E.A.; Zetoune, F.S.; Huber-Lang, M.; Russell, M.W.; Ward, P.A. Complement-induced activation of the cardiac nlrp3 inflammasome in sepsis. FASEB J. 2016, 30, 3997–4006. [Google Scholar] [CrossRef]
  40. Wagner, S.; Schurmann, S.; Hein, S.; Schuttler, J.; Friedrich, O. Septic cardiomyopathy in rat lps-induced endotoxemia: Relative contribution of cellular diastolic ca(2+) removal pathways, myofibrillar biomechanics properties and action of the cardiotonic drug levosimendan. Basic Res. Cardiol. 2015, 110, 507. [Google Scholar] [CrossRef]
  41. Koentges, C.; Cimolai, M.C.; Pfeil, K.; Wolf, D.; Marchini, T.; Tarkhnishvili, A.; Hoffmann, M.M.; Odening, K.E.; Diehl, P.; von Zur Muhlen, C.; et al. Impaired sirt3 activity mediates cardiac dysfunction in endotoxemia by calpain-dependent disruption of atp synthesis. J. Mol. Cell. Cardiol. 2019, 133, 138–147. [Google Scholar] [CrossRef] [PubMed]
  42. Luo, Y.; Fan, C.; Yang, M.; Dong, M.; Bucala, R.; Pei, Z.; Zhang, Y.; Ren, J. Cd74 knockout protects against lps-induced myocardial contractile dysfunction through ampk-skp2-suv39h1-mediated demethylation of bclb. Br. J. Pharm. 2020, 177, 1881–1897. [Google Scholar] [CrossRef] [PubMed]
  43. Joseph, L.C.; Reyes, M.V.; Lakkadi, K.R.; Gowen, B.H.; Hasko, G.; Drosatos, K.; Morrow, J.P. Pkcdelta causes sepsis-induced cardiomyopathy by inducing mitochondrial dysfunction. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H778–H786. [Google Scholar] [CrossRef] [PubMed]
  44. Hobai, I.A.; Edgecomb, J.; LaBarge, K.; Colucci, W.S. Dysregulation of intracellular calcium transporters in animal models of sepsis-induced cardiomyopathy. Shock 2015, 43, 3–15. [Google Scholar] [CrossRef]
  45. DeGrande, S.T.; Little, S.C.; Nixon, D.J.; Wright, P.; Snyder, J.; Dun, W.; Murphy, N.; Kilic, A.; Higgins, R.; Binkley, P.F.; et al. Molecular mechanisms underlying cardiac protein phosphatase 2a regulation in heart. J. Biol. Chem. 2013, 288, 1032–1046. [Google Scholar] [CrossRef] [PubMed]
  46. Antonucci, E.; Fiaccadori, E.; Donadello, K.; Taccone, F.S.; Franchi, F.; Scolletta, S. Myocardial depression in sepsis: From pathogenesis to clinical manifestations and treatment. J. Crit. Care 2014, 29, 500–511. [Google Scholar] [CrossRef]
  47. Zaky, A.; Deem, S.; Bendjelid, K.; Treggiari, M.M. Characterization of cardiac dysfunction in sepsis: An ongoing challenge. Shock 2014, 41, 12–24. [Google Scholar] [CrossRef]
  48. Vallejo, J.G. Role of toll-like receptors in cardiovascular diseases. Clin. Sci. 2011, 121, 1–10. [Google Scholar] [CrossRef]
  49. Yang, J.; Zhang, R.; Jiang, X.; Lv, J.; Li, Y.; Ye, H.; Liu, W.; Wang, G.; Zhang, C.; Zheng, N.; et al. Toll-like receptor 4-induced ryanodine receptor 2 oxidation and sarcoplasmic reticulum ca(2+) leakage promote cardiac contractile dysfunction in sepsis. J. Biol. Chem. 2018, 293, 794–807. [Google Scholar] [CrossRef]
  50. Nath, K.A.; Belcher, J.D.; Nath, M.C.; Grande, J.P.; Croatt, A.J.; Ackerman, A.W.; Katusic, Z.S.; Vercellotti, G.M. Role of tlr4 signaling in the nephrotoxicity of heme and heme proteins. Am. J. Physiol. Ren. Physiol. 2018, 314, F906–F914. [Google Scholar] [CrossRef]
  51. Fattahi, F.; Frydrych, L.M.; Bian, G.; Kalbitz, M.; Herron, T.J.; Malan, E.A.; Delano, M.J.; Ward, P.A. Role of complement c5a and histones in septic cardiomyopathy. Mol. Immunol. 2018, 102, 32–41. [Google Scholar] [CrossRef] [PubMed]
  52. Raeburn, C.D.; Calkins, C.M.; Zimmerman, M.A.; Song, Y.; Ao, L.; Banerjee, A.; Harken, A.H.; Meng, X. Icam-1 and vcam-1 mediate endotoxemic myocardial dysfunction independent of neutrophil accumulation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2002, 283, R477–R486. [Google Scholar] [CrossRef] [PubMed]
  53. Rastaldo, R.; Pagliaro, P.; Cappello, S.; Penna, C.; Mancardi, D.; Westerhof, N.; Losano, G. Nitric oxide and cardiac function. Life Sci. 2007, 81, 779–793. [Google Scholar] [CrossRef]
  54. Kakihana, Y.; Ito, T.; Nakahara, M.; Yamaguchi, K.; Yasuda, T. Sepsis-induced myocardial dysfunction: Pathophysiology and management. J. Intensive Care 2016, 4, 22. [Google Scholar] [CrossRef] [PubMed]
  55. Spiller, F.; Oliveira Formiga, R.; Fernandes da Silva Coimbra, J.; Alves-Filho, J.C.; Cunha, T.M.; Cunha, F.Q. Targeting nitric oxide as a key modulator of sepsis, arthritis and pain. Nitric Oxide 2019, 89, 32–40. [Google Scholar] [CrossRef] [PubMed]
  56. Khadour, F.H.; Panas, D.; Ferdinandy, P.; Schulze, C.; Csont, T.; Lalu, M.M.; Wildhirt, S.M.; Schulz, R. Enhanced no and superoxide generation in dysfunctional hearts from endotoxemic rats. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H1108–H1115. [Google Scholar] [CrossRef] [PubMed]
  57. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [PubMed]
  58. Kirov, M.Y.; Evgenov, O.V.; Evgenov, N.V.; Egorina, E.M.; Sovershaev, M.A.; Sveinbjørnsson, B.; Nedashkovsky, E.V.; Bjertnaes, L.J. Infusion of methylene blue in human septic shock: A pilot, randomized, controlled study. Crit. Care Med. 2001, 29, 1860–1867. [Google Scholar] [CrossRef]
  59. Hollenberg, S.M.; Cunnion, R.E.; Zimmerberg, J. Nitric oxide synthase inhibition reverses arteriolar hyporesponsiveness to catecholamines in septic rats. Am. J. Physiol. 1993, 264, H660–H663. [Google Scholar] [CrossRef]
  60. Rudiger, A.; Dyson, A.; Felsmann, K.; Carré, J.E.; Taylor, V.; Hughes, S.; Clatworthy, I.; Protti, A.; Pellerin, D.; Lemm, J.; et al. Early functional and transcriptomic changes in the myocardium predict outcome in a long-term rat model of sepsis. Clin. Sci. 2013, 124, 391–401. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, Y.C.; Yu, M.M.; Shou, S.T.; Chai, Y.F. Sepsis-induced cardiomyopathy: Mechanisms and treatments. Front. Immunol. 2017, 8, 1021. [Google Scholar] [CrossRef]
  62. Rehman, A.; Baloch, N.U.; Morrow, J.P.; Pacher, P.; Haskó, G. Targeting of g-protein coupled receptors in sepsis. Pharmacol. Ther. 2020, 211, 107529. [Google Scholar] [CrossRef]
  63. Celes, M.R.; Malvestio, L.M.; Suadicani, S.O.; Prado, C.M.; Figueiredo, M.J.; Campos, E.C.; Freitas, A.C.; Spray, D.C.; Tanowitz, H.B.; da Silva, J.S.; et al. Disruption of calcium homeostasis in cardiomyocytes underlies cardiac structural and functional changes in severe sepsis. PLoS ONE 2013, 8, e68809. [Google Scholar] [CrossRef]
  64. Macarthur, H.; Westfall, T.C.; Riley, D.P.; Misko, T.P.; Salvemini, D. Inactivation of catecholamines by superoxide gives new insights on the pathogenesis of septic shock. Proc. Natl. Acad. Sci. USA 2000, 97, 9753–9758. [Google Scholar]
  65. Kalbitz, M.; Fattahi, F.; Herron, T.J.; Grailer, J.J.; Jajou, L.; Lu, H.; Huber-Lang, M.; Zetoune, F.S.; Sarma, J.V.; Day, S.M.; et al. Complement destabilizes cardiomyocyte function in vivo after polymicrobial sepsis and in vitro. J. Immunol. 2016, 197, 2353–2361. [Google Scholar] [CrossRef]
  66. Nuding, S.; Schröder, J.; Presek, P.; Wienke, A.; Müller-Werdan, U.; Ebelt, H.; Werdan, K. Reducing elevated heart rates in patients with multiple organ dysfunction syndrome with the if (funny channel current) inhibitor ivabradine. Shock 2018, 49, 402–411. [Google Scholar] [CrossRef]
  67. Dal-Secco, D.; DalBó, S.; Lautherbach, N.E.S.; Gava, F.N.; Celes, M.R.N.; Benedet, P.O.; Souza, A.H.; Akinaga, J.; Lima, V.; Silva, K.P.; et al. Cardiac hyporesponsiveness in severe sepsis is associated with nitric oxide-dependent activation of g protein receptor kinase. Am. J. Physiol. Heart Circ. Physiol. 2017, 313, H149–H163. [Google Scholar] [CrossRef]
  68. 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]
  69. Ince, C.; Mayeux, P.R.; Nguyen, T.; Gomez, H.; Kellum, J.A.; Ospina-Tascón, G.A.; Hernandez, G.; Murray, P.; De Backer, D. The endothelium in sepsis. Shock 2016, 45, 259–270. [Google Scholar] [CrossRef]
  70. Liu, S.F.; Newton, R.; Evans, T.W.; Barnes, P.J. Differential regulation of cyclo-oxygenase-1 and cyclo-oxygenase-2 gene expression by lipopolysaccharide treatment in vivo in the rat. Clin. Sci. 1996, 90, 301–306. [Google Scholar] [CrossRef]
  71. Zidar, N.; Dolenc-Strazar, Z.; Jeruc, J.; Jerse, M.; Balazic, J.; Gartner, U.; Jermol, U.; Zupanc, T.; Stajer, D. Expression of cyclooxygenase-1 and cyclooxygenase-2 in the normal human heart and in myocardial infarction. Cardiovasc. Pathol. 2007, 16, 300–304. [Google Scholar] [CrossRef]
  72. Reines, H.D.; Halushka, P.V.; Cook, J.A.; Wise, W.C.; Rambo, W. Plasma thromboxane concentrations are raised in patients dying with septic shock. Lancet 1982, 2, 174–175. [Google Scholar] [CrossRef]
  73. Stewart, D.J.; Cernacek, P.; Costello, K.B.; Rouleau, J.L. Elevated endothelin-1 in heart failure and loss of normal response to postural change. Circulation 1992, 85, 510–517. [Google Scholar] [CrossRef]
  74. Freeman, B.D.; Machado, F.S.; Tanowitz, H.B.; Desruisseaux, M.S. Endothelin-1 and its role in the pathogenesis of infectious diseases. Life Sci. 2014, 118, 110–119. [Google Scholar] [CrossRef]
  75. Yang, L.L.; Gros, R.; Kabir, M.G.; Sadi, A.; Gotlieb, A.I.; Husain, M.; Stewart, D.J. Conditional cardiac overexpression of endothelin-1 induces inflammation and dilated cardiomyopathy in mice. Circulation 2004, 109, 255–261. [Google Scholar] [CrossRef]
  76. Carrara, M.; Ferrario, M.; Bollen Pinto, B.; Herpain, A. The autonomic nervous system in septic shock and its role as a future therapeutic target: A narrative review. Ann. Intensive Care 2021, 11, 80. [Google Scholar] [CrossRef]
  77. Fillmore, N.; Mori, J.; Lopaschuk, G.D. Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br. J. Pharm. 2014, 171, 2080–2090. [Google Scholar] [CrossRef]
  78. Tessier, J.P.; Thurner, B.; Jüngling, E.; Lückhoff, A.; Fischer, Y. Impairment of glucose metabolism in hearts from rats treated with endotoxin. Cardiovasc. Res. 2003, 60, 119–130. [Google Scholar] [CrossRef]
  79. Memon, R.A.; Fuller, J.; Moser, A.H.; Smith, P.J.; Feingold, K.R.; Grunfeld, C. In vivo regulation of acyl-coa synthetase mrna and activity by endotoxin and cytokines. Am. J. Physiol. 1998, 275, E64–E72. [Google Scholar] [CrossRef]
  80. Memon, R.A.; Bass, N.M.; Moser, A.H.; Fuller, J.; Appel, R.; Grunfeld, C.; Feingold, K.R. Down-regulation of liver and heart specific fatty acid binding proteins by endotoxin and cytokines in vivo. Biochim. Biophys. Acta 1999, 1440, 118–126. [Google Scholar] [CrossRef]
  81. Drosatos, K.; Khan, R.S.; Trent, C.M.; Jiang, H.; Son, N.H.; Blaner, W.S.; Homma, S.; Schulze, P.C.; Goldberg, I.J. Peroxisome proliferator-activated receptor-γ activation prevents sepsis-related cardiac dysfunction and mortality in mice. Circ. Heart Fail. 2013, 6, 550–562. [Google Scholar] [CrossRef] [Green Version]
  82. Brealey, D.; Brand, M.; Hargreaves, I.; Heales, S.; Land, J.; Smolenski, R.; Davies, N.A.; Cooper, C.E.; Singer, M. Association between mitochondrial dysfunction and severity and outcome of septic shock. Lancet 2002, 360, 219–223. [Google Scholar] [CrossRef]
  83. Stanzani, G.; Duchen, M.R.; Singer, M. The role of mitochondria in sepsis-induced cardiomyopathy. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 759–773. [Google Scholar] [CrossRef]
  84. Brealey, D.; Karyampudi, S.; Jacques, T.S.; Novelli, M.; Stidwill, R.; Taylor, V.; Smolenski, R.T.; Singer, M. Mitochondrial dysfunction in a long-term rodent model of sepsis and organ failure. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 286, R491–R497. [Google Scholar] [CrossRef]
  85. Hotchkiss, R.S.; Nicholson, D.W. Apoptosis and caspases regulate death and inflammation in sepsis. Nat. Rev. Immunol. 2006, 6, 813–822. [Google Scholar] [CrossRef]
  86. Crouser, E.D. Mitochondrial dysfunction in septic shock and multiple organ dysfunction syndrome. Mitochondrion 2004, 4, 729–741. [Google Scholar] [CrossRef]
  87. Dikalov, S. Cross talk between mitochondria and nadph oxidases. Free Radic. Biol. Med. 2011, 51, 1289–1301. [Google Scholar] [CrossRef]
  88. Bernardi, P.; Di Lisa, F. The mitochondrial permeability transition pore: Molecular nature and role as a target in cardioprotection. J. Mol. Cell. Cardiol. 2015, 78, 100–106. [Google Scholar] [CrossRef]
  89. Turdi, S.; Han, X.; Huff, A.F.; Roe, N.D.; Hu, N.; Gao, F.; Ren, J. Cardiac-specific overexpression of catalase attenuates lipopolysaccharide-induced myocardial contractile dysfunction: Role of autophagy. Free Radic. Biol. Med. 2012, 53, 1327–1338. [Google Scholar] [CrossRef]
  90. Torraco, A.; Carrozzo, R.; Piemonte, F.; Pastore, A.; Tozzi, G.; Verrigni, D.; Assenza, M.; Orecchioni, A.; D’Egidio, A.; Marraffa, E.; et al. Effects of levosimendan on mitochondrial function in patients with septic shock: A randomized trial. Biochimie 2014, 102, 166–173. [Google Scholar] [CrossRef]
  91. Matkovich, S.J.; Al Khiami, B.; Efimov, I.R.; Evans, S.; Vader, J.; Jain, A.; Brownstein, B.H.; Hotchkiss, R.S.; Mann, D.L. Widespread down-regulation of cardiac mitochondrial and sarcomeric genes in patients with sepsis. Crit. Care Med. 2017, 45, 407–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sevilla Berrios, R.A.; O’Horo, J.C.; Velagapudi, V.; Pulido, J.N. Correlation of left ventricular systolic dysfunction determined by low ejection fraction and 30-day mortality in patients with severe sepsis and septic shock: A systematic review and meta-analysis. J. Crit. Care 2014, 29, 495–499. [Google Scholar] [CrossRef] [PubMed]
  93. Paonessa, J.R.; Brennan, T.; Pimentel, M.; Steinhaus, D.; Feng, M.; Celi, L.A. Hyperdynamic left ventricular ejection fraction in the intensive care unit. Crit. Care 2015, 19, 288. [Google Scholar] [CrossRef] [PubMed]
  94. Sanfilippo, F.; Corredor, C.; Fletcher, N.; Tritapepe, L.; Lorini, F.L.; Arcadipane, A.; Vieillard-Baron, A.; Cecconi, M. Left ventricular systolic function evaluated by strain echocardiography and relationship with mortality in patients with severe sepsis or septic shock: A systematic review and meta-analysis. Crit. Care 2018, 22, 183. [Google Scholar] [CrossRef] [PubMed]
  95. Sanfilippo, F.; Corredor, C.; Arcadipane, A.; Landesberg, G.; Vieillard-Baron, A.; Cecconi, M.; Fletcher, N. Tissue doppler assessment of diastolic function and relationship with mortality in critically ill septic patients: A systematic review and meta-analysis. Br. J. Anaesth. 2017, 119, 583–594. [Google Scholar] [CrossRef]
  96. Kim, J.S.; Kim, Y.J.; Kim, M.; Ryoo, S.M.; Kim, W.Y. Association between right ventricle dysfunction and poor outcome in patients with septic shock. Heart 2020, 106, 1665–1671. [Google Scholar] [CrossRef]
  97. Lanspa, M.J.; Cirulis, M.M.; Wiley, B.M.; Olsen, T.D.; Wilson, E.L.; Beesley, S.J.; Brown, S.M.; Hirshberg, E.L.; Grissom, C.K. Right ventricular dysfunction in early sepsis and septic shock. Chest 2021, 159, 1055–1063. [Google Scholar] [CrossRef]
  98. Vallabhajosyula, S.; Kumar, M.; Pandompatam, G.; Sakhuja, A.; Kashyap, R.; Kashani, K.; Gajic, O.; Geske, J.B.; Jentzer, J.C. Prognostic impact of isolated right ventricular dysfunction in sepsis and septic shock: An 8-year historical cohort study. Ann. Intensive Care 2017, 7, 94. [Google Scholar] [CrossRef]
  99. Dugar, S.P.; Vallabhajosyula, S. Right ventricle in sepsis: Clinical and research priority. Heart 2020, 106, 1629–1630. [Google Scholar] [CrossRef]
  100. Werdan, K.; Oelke, A.; Hettwer, S.; Nuding, S.; Bubel, S.; Hoke, R.; Russ, M.; Lautenschlager, C.; Mueller-Werdan, U.; Ebelt, H. Septic cardiomyopathy: Hemodynamic quantification, occurrence, and prognostic implications. Clin. Res. Cardiol. 2011, 100, 661–668. [Google Scholar] [CrossRef]
  101. Chen, W.Y.; Zhang, Z.H.; Tao, L.L.; Xu, Q.; Wei, X.; Chen, M.S. Afterload-related cardiac performance identifies cardiac impairment and associates with outcome in patients with septic shock: A retrospective cohort study. J. Intensive Care 2021, 9, 33. [Google Scholar] [CrossRef] [PubMed]
  102. Zhao, C.C.; Zhang, L.R.; Liu, L.X.; Sun, L.X.; Hu, Z.J. Afterload-related cardiac performance predicts prognosis in critical ill patients with sepsis: A prospective observational pilot study. Medicine 2021, 100, e27235. [Google Scholar] [CrossRef] [PubMed]
  103. Wilhelm, J.; Hettwer, S.; Schuermann, M.; Bagger, S.; Gerhardt, F.; Mundt, S.; Muschik, S.; Zimmermann, J.; Bubel, S.; Amoury, M.; et al. Severity of cardiac impairment in the early stage of community-acquired sepsis determines worse prognosis. Clin. Res. Cardiol. 2013, 102, 735–744. [Google Scholar] [CrossRef] [PubMed]
  104. Jeong, H.S.; Lee, T.H.; Bang, C.H.; Kim, J.H.; Hong, S.J. Risk factors and outcomes of sepsis-induced myocardial dysfunction and stress-induced cardiomyopathy in sepsis or septic shock: A comparative retrospective study. Medicine 2018, 97, e0263. [Google Scholar] [CrossRef]
  105. Landesberg, G.; Jaffe, A.S.; Gilon, D.; Levin, P.D.; Goodman, S.; Abu-Baih, A.; Beeri, R.; Weissman, C.; Sprung, C.L.; Landesberg, A. Troponin elevation in severe sepsis and septic shock: The role of left ventricular diastolic dysfunction and right ventricular dilatation. Crit. Care Med. 2014, 42, 790–800. [Google Scholar] [CrossRef]
  106. Pandompatam, G.; Kashani, K.; Vallabhajosyula, S. The role of natriuretic peptides in the management, outcomes and prognosis of sepsis and septic shock. Rev. Bras. Ter. Intensiva 2019, 31, 368–378. [Google Scholar] [CrossRef]
  107. Bai, Y.L.; Hu, B.L.; Wen, H.C.; Zhang, Y.L.; Zhu, J.J. Prognostic value of plasma brain natriuretic peptide value for patientswith sepsis: A meta-analysis. J. Crit. Care 2018, 48, 145–152. [Google Scholar] [CrossRef]
  108. Vallabhajosyula, S.; Wang, Z.; Murad, M.H.; Vallabhajosyula, S.; Sundaragiri, P.R.; Kashani, K.; Miller, W.L.; Jaffe, A.S.; Vallabhajosyula, S. Natriuretic peptides to predict short-term mortality in patients with sepsis: A systematic review and meta-analysis. Mayo Clin. Proc. Innov. Qual. Outcomes 2020, 4, 50–64. [Google Scholar] [CrossRef]
  109. Beltran-Garcia, J.; Osca-Verdegal, R.; Nacher-Sendra, E.; Cardona-Monzonis, A.; Sanchis-Gomar, F.; Carbonell, N.; Pallardo, F.V.; Lavie, C.J.; Garcia-Gimenez, J.L. Role of non-coding rnas as biomarkers of deleterious cardiovascular effects in sepsis. Prog. Cardiovasc. Dis. 2021, 68, 70–77. [Google Scholar] [CrossRef]
  110. Chen, X.; Liu, X.; Dong, R.; Zhang, D.; Qin, S. A retrospective observational study of the association between plasma levels of interleukin 8 in 42 patients with sepsis-induced myocardial dysfunction at a single center between 2017 and 2020. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2021, 27, e933065. [Google Scholar] [CrossRef]
  111. Chen, M.; Kong, C.; Zheng, Z.; Li, Y. Identification of biomarkers associated with septic cardiomyopathy based on bioinformatics analyses. J. Comput. Biol. A J. Comput. Mol. Cell Biol. 2020, 27, 69–80. [Google Scholar] [CrossRef] [PubMed]
  112. Pro, C.I.; Yealy, D.M.; Kellum, J.A.; Huang, D.T.; Barnato, A.E.; Weissfeld, L.A.; Pike, F.; Terndrup, T.; Wang, H.E.; Hou, P.C.; et al. A randomized trial of protocol-based care for early septic shock. N. Engl. J. Med. 2014, 370, 1683–1693. [Google Scholar]
  113. Investigators, A.; Group, A.C.T.; Peake, S.L.; Delaney, A.; Bailey, M.; Bellomo, R.; Cameron, P.A.; Cooper, D.J.; Higgins, A.M.; Holdgate, A.; et al. Goal-directed resuscitation for patients with early septic shock. N. Engl. J. Med. 2014, 371, 1496–1506. [Google Scholar]
  114. Mouncey, P.R.; Osborn, T.M.; Power, G.S.; Harrison, D.A.; Sadique, M.Z.; Grieve, R.D.; Jahan, R.; Harvey, S.E.; Bell, D.; Bion, J.F.; et al. Trial of early, goal-directed resuscitation for septic shock. N. Engl. J. Med. 2015, 372, 1301–1311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Diagnostic challenges in septic cardiomyopathy are a consequence of its complex pathophysiology. Recent advances in cardiac imaging paradoxically increased the uncertainty on how to define sepsis-induced cardiomyopathy. Underlying myocardial dysfunction, heart rate and the need of acute interventions (i.e., fluid challenge and inotrope use) add further layer of complexity. These issues highlight the importance of a dynamic assessment of cardiac performance over time, not limited to echocardiographic findings but also considering the cardiac performance.
Figure 1. Diagnostic challenges in septic cardiomyopathy are a consequence of its complex pathophysiology. Recent advances in cardiac imaging paradoxically increased the uncertainty on how to define sepsis-induced cardiomyopathy. Underlying myocardial dysfunction, heart rate and the need of acute interventions (i.e., fluid challenge and inotrope use) add further layer of complexity. These issues highlight the importance of a dynamic assessment of cardiac performance over time, not limited to echocardiographic findings but also considering the cardiac performance.
Cells 11 02833 g001
Figure 2. Hemodynamic stress and systemic inflammation exert a synergistic effect on myocardial function impairment during sepsis. Systemic response to sepsis triggers and sustains myocardial dysfunction through several pathways: by impairing the compensatory reaction of sympathetic system with consequent microvascular dysfunction and—In parallel—through theimmune system activation and microvascular dysfunction (black arrows). Those mechanisms deeply impair myocardial tissue through a cascade involving damage- and pathogen-associated molecular pattern (DAMPs and PAMPs, respectively), Toll-like receptors (TLRs), complement and release of cytokines and reactive oxygen species.
Figure 2. Hemodynamic stress and systemic inflammation exert a synergistic effect on myocardial function impairment during sepsis. Systemic response to sepsis triggers and sustains myocardial dysfunction through several pathways: by impairing the compensatory reaction of sympathetic system with consequent microvascular dysfunction and—In parallel—through theimmune system activation and microvascular dysfunction (black arrows). Those mechanisms deeply impair myocardial tissue through a cascade involving damage- and pathogen-associated molecular pattern (DAMPs and PAMPs, respectively), Toll-like receptors (TLRs), complement and release of cytokines and reactive oxygen species.
Cells 11 02833 g002
Table 1. Definition of septic cardiomyopathy.
Table 1. Definition of septic cardiomyopathy.
RefsPhenotypesDefining Criteria
Diagnostic criteria for sepsis-associated cardiac dysfunction unrelated to ischemia *.
Beesley et al., 2018 [5]
Martin at al., 2019 [6]
(i)
LV dilatation with normal- or low-filling pressure
NA
(ii)
Reduced LV ventricular contractility
LVEF < 40–50%, ↓ LVFAC
(iii)
RV dysfunction of LV (systolic or diastolic) dysfunction with reduce response to fluid challenge
LVEF < 40–50%, ↓ LVFAC, e’ velocity, MPI, Afterload-related cardiac performance, Ventricular arterial decoupling
Cardiovascular phenotypes of septic cardiomyopathy classification performance
Geri et al., 2019 [7]
(i)
well resuscitated without myocardial dysfunction or fluid responsiveness
(ii)
LV systolic dysfunction
LVEF < 40%, LVFAC < 33%, aortic VTI < 14 cm
(iii)
Hyperkinetic profile
Aortic VTI > 20 cm, LVFAC > 58%, heart rate < 106 bpm
(iv)
RV failure
RV/LV EDA > 0.8, sBP < 100 mmHg, dBP < 51 mmHg
(v)
persistent hypovolemia
Aortic VTI < 16 cm, E wave < 67 cm/s, ΔDSVC > 39%
* One or more of these characteristics may define septic cardiomyopathy. LV: left ventricular; LVEF: left ventricular ejection fraction; LVFAC: left ventricular fractional area change; RV: right ventricle; MPI: myocardial performance index; MVG: myocardial velocity gradient; STE: speckle tracking echocardiography; VTI: velocity-time integral; EDA: end-diastolic area; sBP: systolic blood pressure; dBP: diastolic blood pressure; ΔSVC: respiratory variations of the superior vena cava.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Carbone, F.; Liberale, L.; Preda, A.; Schindler, T.H.; Montecucco, F. Septic Cardiomyopathy: From Pathophysiology to the Clinical Setting. Cells 2022, 11, 2833. https://doi.org/10.3390/cells11182833

AMA Style

Carbone F, Liberale L, Preda A, Schindler TH, Montecucco F. Septic Cardiomyopathy: From Pathophysiology to the Clinical Setting. Cells. 2022; 11(18):2833. https://doi.org/10.3390/cells11182833

Chicago/Turabian Style

Carbone, Federico, Luca Liberale, Alberto Preda, Thomas Hellmut Schindler, and Fabrizio Montecucco. 2022. "Septic Cardiomyopathy: From Pathophysiology to the Clinical Setting" Cells 11, no. 18: 2833. https://doi.org/10.3390/cells11182833

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