Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML

  • Supplementary Files
    • Supplementary_Data.pdf
Review Open Access

Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)

  • Authors:
    • Fei Tang
    • Dong Liu
    • Shi-Chao Zhu
    • Hui-Min Zhou
    • Xue-Wen Qiu
  • View Affiliations / Copyright

    Affiliations: Department of Pharmacy, Chongqing General Hospital, Chongqing University, Chongqing 400014, P.R. China, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
    Copyright: © Tang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 71
    |
    Published online on: January 22, 2026
       https://doi.org/10.3892/ijmm.2026.5742
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:


Abstract

Sepsis, an infection‑triggered systemic inflammatory response syndrome, ranks as the third leading cause of death worldwide due to its high incidence and mortality. Sepsis‑induced myocardial dysfunction (SIMD) is a frequent and serious complication that notably increases patient morbidity and mortality. The underlying pathophysiology of SIMD involves a complex interplay of inflammation, oxidative stress, mitochondrial impairment and apoptosis, yet no effective therapies have been established. Thus, uncovering the molecular mechanisms of SIMD, identifying novel therapeutic targets and developing efficacious agents are key. For centuries, natural products have been used in traditional medical systems across China and Asia to manage cardiovascular disease. These compounds can confer cardioprotection by modulating inflammatory pathways, decreasing oxidative stress, inhibiting apoptotic cell death and improving mitochondrial function. The present review aimed to summarize the clinical manifestations and pathophysiology of SIMD and how natural products exert their protective effects. The present study aimed to explore structure‑activity relationships and highlight key molecular targets and representative natural product binding affinities for SIMD‑related proteins. In summary, the present study presents a comprehensive overview of the multi‑targeted strategies employed by natural products against SIMD and provides guidance for the discovery of SIMD‑focused dietary supplements and lead compounds, laying the groundwork for future translational research.

Introduction

Sepsis, an infection-driven systemic inflammatory response syndrome, poses a major global health challenge due to its high incidence and mortality (1,2). According to World Health Organization estimates, nearly 19 million people develop sepsis annually and 20-30% succumb to severe complications (2). In intensive care units, mortality is up to 56%, making sepsis the third leading cause of death worldwide (3,4). Among complications, sepsis-induced myocardial dysfunction (SIMD) is especially common and carries the worst prognosis. A total of ~70% of septic patients experience impaired myocardial contractility, decreased ejection fraction and arrhythmia. These cardiac impairments often coincide with hypotension and lactic acidosis, hastening circulatory collapse and multi-organ failure (5-7).

SIMD arises from a multifactorial network of inflammatory and oxidative processes. Pathogen-associated molecular patterns (PAMPs) engage toll-like receptor (TLR)4 signaling through MyD88 and TIR-domain-containing adaptor inducing interferon-β, thereby activating NF-κB and MAPKs (p38, JNK, ERK). This cascade triggers the release of proinflammatory cytokines such as TNF-α and IL-1β (8). The cytokine surge, in turn, stimulates NADPH oxidase to generate reactive oxygen species (ROS), which induce lipid peroxidation and protein damage (9). Oxidative stress further compromises mitochondrial integrity, leading to decreased ATP synthesis and the activation of caspase-3-mediated apoptosis. Collectively, these events depress ejection fraction, promote ventricular dilation and drive circulatory failure (8).

Current clinical management of SIMD follows the Surviving Sepsis Campaign Guidelines, focusing on fluid resuscitation, broad-spectrum antibiotics and vasopressors. Inotropes or β-blockers may be used as adjunctive therapies, but they typically yield only transient hemodynamic benefits and can increase myocardial oxygen demand or provoke arrhythmia (5). Given the intertwined inflammation-oxidation-mitochondria-apoptosis network in SIMD, single-target drugs are unlikely to provide comprehensive cardioprotection. However, natural products offer promising multi-target interventions with low toxicity profiles (10). Traditional compounds such as flavonoids, glycosides, alkaloids and saponins exhibit anti-inflammatory, antioxidant, antiapoptotic and mitochondrial-restorative effects. Preclinical study demonstrates their capacity to attenuate key aspects of SIMD pathology (10); for example, curcumin has been found to have extensive cardiovascular protective effects by regulating endothelial function (10).

To the best of our knowledge, a systematic synthesis of how natural products counteract SIMD is lacking. The present review therefore summarizes the clinical features and pathophysiology of SIMD and the structure-activity-mechanism relationships of major natural product classes. Furthermore, the present study aimed to highlight the primary signaling pathways involved and evaluate advanced delivery strategies that improve bioavailability and representative compound binding to SIMD targets. By offering an integrated framework, the present study aimed to guide the development of novel multi-target therapeutic agents, including dietary supplements and lead compounds, for SIMD therapy.

Pathophysiology of SIMD

SIMD is a complex, dynamic process in which overwhelming infection and systemic inflammation disrupt normal cardiac function. Multiple interrelated mechanisms, including dysregulated cytokine release, oxidative stress, mitochondrial damage and impaired calcium handling, converge to depress myocardial performance (5). A comprehensive understanding of these pathways is key for accurate diagnosis and designing targeted therapies (Fig. 1).

Pathophysiological mechanisms of
SIMD. This schematic illustrates the key interrelated pathways
(such as PI3K/Akt, NLRP3 and NF-κB) contributing to SIMD, including
dysregulated cytokine release, oxidative stress, mitochondrial
damage, impaired calcium handling and the regulatory roles of
specific. miRs. SIMD, sepsis-induced myocardial dysfunction; miR,
microRNA HMGB, high mobility group box; TLRS, Toll like receptors;
ROS, reactive oxygen species; SOD, superoxide dismutase; MDA,
malondialdehyde; iNOS, inducible nitric oxide synthase; RNS,
reactive nitrogen species; DRP, dynamin-related protein; PPAR,
peroxisome proliferator activated-receptors; mPTP, mitochondrial
permeability transition pore.

Figure 1

Pathophysiological mechanisms of SIMD. This schematic illustrates the key interrelated pathways (such as PI3K/Akt, NLRP3 and NF-κB) contributing to SIMD, including dysregulated cytokine release, oxidative stress, mitochondrial damage, impaired calcium handling and the regulatory roles of specific. miRs. SIMD, sepsis-induced myocardial dysfunction; miR, microRNA HMGB, high mobility group box; TLRS, Toll like receptors; ROS, reactive oxygen species; SOD, superoxide dismutase; MDA, malondialdehyde; iNOS, inducible nitric oxide synthase; RNS, reactive nitrogen species; DRP, dynamin-related protein; PPAR, peroxisome proliferator activated-receptors; mPTP, mitochondrial permeability transition pore.

Clinical characteristics of SIMD

SIMD describes a transient, sepsis-associated decline in myocardial function that can involve both the left and right ventricles and may affect systolic and/or diastolic performance (11). Many studies define SIMD by a reversible decrease in left ventricular ejection fraction (LVEF) accompanied by ventricular dilation and poor response to fluid resuscitation or catecholamines (3,5,7). However, because LVEF is load-dependent, it may not reliably reflect contractile reserve during sepsis (12). Consequently, SIMD is now recognized as a spectrum of load-independent myocardial depression manifesting as left ventricular systolic or diastolic dysfunction, right ventricular dysfunction or a combination of both, often with fluctuating hemodynamics (13).

Clinically, SIMD typically emerges within the first hours to days of septic illness and presents with a heterogeneous cardiac phenotype (14). Systolic abnormalities range from decreased contractility to ventricular dilation in certain patients (15), while diastolic dysfunction is identified by altered filling parameters. Right ventricular involvement is not uncommon and may exacerbate hemodynamic instability (16). SIMD is often reversible, although persistent dysfunction and worse outcomes occur depending on sepsis severity, underlying comorbidities and the level of hemodynamic support required (17). The clinical presentation may evolve over time, with an initial high-output state potentially progressing to a low-output phase, reflecting the interplay between preload, afterload, myocardial depression and microcirculatory redistribution (18). Thus, in clinical practice, the diagnosis of SIMD relies on a comprehensive, multi-faceted assessment. The typically involves confirming the presence of sepsis, identifying otherwise unexplained myocardial dysfunction, evidenced by new and often reversible echocardiographic abnormality and associating these findings with clinical signs of hemodynamic compromise, such as persistent vasopressor dependency or objective evidence of tissue hypoperfusion (14). While elevated biomarkers such as cardiac troponins and natriuretic peptides support the diagnosis by indicating myocardial injury or stress, they are not standalone diagnostic criteria due to their lack of specificity in the septic context (14).

Biomarkers of SIMD

Biomarkers of SIMD reflect diverse pathophysiological domains, including direct myocardial injury, ventricular wall stress, inflammatory remodeling and systemic perfusion. It is crucial to note that most currently used biomarkers are not specific to SIMD but indicate general myocardial injury, stress, inflammation, or dysfunction. High-sensitivity troponins I and T levels frequently rise in septic patients with cardiac involvement, signaling myocardial injury rather than an acute coronary syndrome. Troponin levels are associated with illness severity and adverse outcomes (12,19), but their release in sepsis is multifactorial, including demand ischemia, microvascular injury, cytokine-mediated toxicity and catecholamine effects, underscoring their lack of specificity for SIMD (19,20). B-type natriuretic peptides (BNP) and N-terminal pro-B-type natriuretic peptide (NT-proBNP) increase with myocardial wall stress and diastolic dysfunction and also predict higher mortality (21,22). However, their variable association with LVEF reduces their value as standalone markers of intrinsic myocardial depression in SIMD (23).

Inflammatory and remodeling markers add prognostic information beyond troponin and NPs. Soluble ST2 and galectin-3 rise in septic patients with myocardial involvement and are associated with severity and adverse outcomes, reflecting ongoing inflammation and fibrosis (24). Similar to other markers, they are not specific to SIMD, and their interpretation requires integration with imaging and clinical context. Early injury markers such as heart-type fatty acid-binding protein (H-FABP) may increase before troponin in certain cases, offering potential for early risk stratification (25). Metabolic and perfusion markers, including lactate, indicate global tissue hypoperfusion and shock severity and thus complement cardiac-specific biomarkers in a comprehensive hemodynamic assessment (26). Specific circulating microRNAs (miRNAs or miRs) implicated in inflammatory and apoptotic pathways (such as miR-21, miR-155 and miR-146a) represent a promising class of biomarkers. They hold potential for more precise SIMD detection and risk stratification but require prospective validation before routine clinical use (27,28).

In summary, a multimodal strategy that integrates biomarkers with load-independent echocardiographic measures and detailed hemodynamic data is the most robust approach to defining myocardial involvement, monitoring progression and guiding management.

Pathology of SIMD
Programmed cell death

Cardiomyocyte loss in sepsis occurs through multiple interrelated programmed cell death pathways (29). Apoptosis proceeds through intrinsic mitochondrial pathways, where Bax and Bak mediate outer mitochondrial membrane permeabilization and caspase activation, and through endoplasmic reticulum stress pathways involving glucose-regulated protein 78 kDa, CHOP and caspase-12 (30-32). Death receptor signaling via TNF-R1 and FAS also contributes to caspase-8-mediated apoptosis. Pharmacological modulation of these signals modestly improves cardiac function in CLP (Cecum ligation and puncture (CLP)-induced rats (33). Necroptosis, driven by receptor interacting serine/threonine kinase RIPK1, RIPK3 and MLKL (Mixed lineage kinase domain-like) MLKL, amplifies inflammation and damages cardiomyocytes; inhibiting RIPK or MLKL attenuates myocardial injury in CLP-induced septic mice (34). Pyroptosis, a lytic form of cell death dependent on caspase-1-mediated gasdermin D (GSDMD) pore formation, releases IL-1β and IL-18 and amplifies systemic inflammation (35,36). Ferroptosis, an iron-dependent form of lipid peroxidation, also contributes to cardiomyocyte death; iron chelators and ferroptosis inhibitors lower ROS and lipid peroxidation to preserve cardiac function in endotoxemia (37,38). Autophagy plays a dual role in SIMD. Moderate autophagic flux supports cell homeostasis and cardiac resilience, but excessive or insufficient autophagy may worsen injury (39). Pharmacological induction of autophagy can restore homeostasis and improve cardiac outcomes in septic rats (40).

Inflammation

Sepsis releases PAMPs and damage-associated molecular patterns, which activate TLRs on cardiomyocytes, fibroblasts and endothelial cells (ECs) (41). Except for TLR3, most TLR signaling proceeds via the adaptor protein MyD88, activating downstream effectors such as the MAPKs JNK, ERK1/2 and p38, as well as the transcription factor NF-κB (42). This cascade drives production of proinflammatory cytokines (such as IL-1β, IL-6, TNF-α, IL-12, IL-17, IL-18) and chemokines such as CCL2/MCP-1 and CXCL8, which recruit and activate immune cells (13,43-45). Activation of the NLRP3 inflammasome further amplifies IL-1β and IL-18 release, thereby creating an inflammatory milieu that impairs cardiomyocyte contractility by altering autophagy and lysosomal function (46). High mobility group box 1) HMGB1 and extracellular histones released during sepsis impair myocardial performance through TLR-dependent pathways, reinforcing the inflammatory cascade (47). Autonomic dysregulation also interacts with inflammation: Altered adrenergic signaling can modulate cytokine production and cardiomyocyte responsiveness, creating self-perpetuating feedback loops that influence disease progression (48).

Oxidative stress

Alongside inflammation, sepsis triggers a surge of ROS and nitrogen species (RNS) in cardiac tissue (49). ROS originate from electron leak in mitochondrial complexes I-III and from NO-derived species such as peroxynitrite (ONOO) (50). Oxidative and nitrosative stress damage lipids, protein and DNA, deplete antioxidants including superoxide dismutase, catalase and glutathione, and disrupt calcium handling and excitation-contraction coupling (51). NO produced by inducible nitric oxide synthase (iNOS) contributes to myocardial depression, while ONOO− nitrates tyrosine residues and further impairs mitochondrial and contractile protein (52). This redox imbalance shifts signaling toward proinflammatory pathways, creating a cycle that worsens contractile dysfunction (53).

Mitochondrial dysfunction

Mitochondria are key for energy supply and cell survival rate in SIMD (54). Sepsis induces structural changes, swelling and cristae disruption, alongside declines in oxidative phosphorylation and ATP synthesis (55). Excessive mitochondrial fission, driven by the GTPase dynamin-related protein 1 (DRP1), fragments the organelle and worsens energy deficits, whereas promoting fusion or mitophagy can maintain mitochondrial integrity in LPS-induced mice (56). The mitochondrial permeability transition pore (mPTP) opens under calcium overload, oxidative stress and ATP depletion, collapsing membrane potential and releasing cytochrome c (57). Agents that prevent mPTP opening or stimulate mitochondrial biogenesis improve mitochondrial function and cardiac performance in septic rats (58). In SIMD, energy metabolism shifts from fatty acid oxidation to glycolysis, decreasing ATP yield; downregulation of lipid-oxidation regulators such as PPARs and α subunit of peroxisome proliferators activated receptor-γ coactivator-1 (PGC-1α exacerbates energetic failure (58). Additional mitochondrial insults include mitochondrial DNA oxidation and impaired activity of electron transport chain complexes II and IV, limiting ATP production and contractile capacity (59). Restoring mitochondrial integrity with targeted antioxidants and NAD+-boosting strategies offers a promising therapeutic approach in SIMD (60).

Endothelial-myocardial crosstalk

ECs coordinate vascular tone, perfusion and inflammation in the cardiovascular system (61). In sepsis, EC dysfunction transforms the vascular bed into a source of injurious signals that affect adjacent cardiomyocytes, fibroblasts and the microcirculation (62). Under inflammatory and oxidative stress, ECs activate TLRs, triggering NF-κB and MAPK pathways and increasing production of ROS and RNS (63). This pro-oxidant environment impairs myocardial relaxation and contraction and worsens microvascular perfusion. Upregulation of intercellular cell adhesion molecule-1 vascular cell adhesion molecule 1) promotes leukocyte trafficking into the myocardium, while degradation of the glycocalyx increases vascular permeability, edema and microvascular leakage, collectively decreasing oxygen delivery and substrate availability to the heart (64,65). Emerging evidence supports bidirectional communication between ECs and cardiomyocytes: Endothelial-derived signals modulate cardiomyocyte function and energy metabolism, while cardiomyocytes influence endothelial behavior via paracrine pathways and hemodynamic feedback (66). For example, endothelial-derived reactive species and altered NO signaling contribute to calcium handling abnormality and energetic stress in cardiomyocytes (67). Conversely, interventions that dampen endothelial inflammation or NO production yield downstream cardioprotective effects (68,69).

Role of miRNAs

Dysregulated miRNAs serve as key epigenetic regulators and downstream effectors in the pathogenesis of SIMD (70). These small non-coding (nc)RNAs fine-tune gene expression post-transcriptionally, predominantly by modulating central inflammatory and apoptotic pathways.

Numerous miRNAs critically regulate the inflammatory cascade in SIMD, primarily through targeting key components of the NF-κB signaling pathway. Certain miRNAs serve as endogenous negative feedback regulators to attenuate excessive inflammation. For example, miR-146a and miR-125b dampen NF-κB activation and subsequent pro-inflammatory cytokine production by targeting key adaptor proteins IL-1 receptor-associated kinase) and TNF receptor associated factor 6) (71,72). Similarly, miR-25 inhibits the TLR4/NF-κB pathway by directly targeting PTEN (73) and miR-29b-3p suppresses MAPK/NF-κB signaling by targeting FOXO3A (74). miR-335, although upregulated in SIMD, appears to exert a net beneficial effect on inflammation and injury when overexpressed, suggesting a complex, context-dependent regulatory role (75). Conversely, specific miRNAs exacerbate myocardial inflammation. miR-193a, expression of which is enhanced via METTL3-mediated m6A modification, promotes inflammation by targeting the anti-apoptotic gene BCL2L2 (76). The circROCK1/miR-96-5p/oxidative stress responsive kinase 1) pathway also promotes myocardial injury and NF-κB activation (77). Furthermore, hsa-miR-23a-3p, hsa-miR-3175 and hsa-miR-23b-3p are implicated in regulating NF-κB signaling via histone deacetylase 7)/ACTN4 (A-actinin-4), contributing to inflammatory damage (78).

Beyond inflammation, miRNAs determine cardiomyocyte fate by fine-tuning apoptotic and other cell death pathways. Multiple miRNAs confer cardioprotection by inhibiting pro-apoptotic signals or enhancing survival pathways. These include miR-214, which improves cardiac function and suppresses apoptosis (79). miR-21 inhibits apoptosis by targeting programmed cell death 4) to activate the PI3K/Akt pathway, as well as by directly regulating Bcl-2 and CDK6 (80). miR-150-5p alleviates apoptosis by modulating Akt2 expression (80). miR-107 promotes cardiomyocyte proliferation and inhibits apoptosis via the PTEN/PI3K/AKT pathway (81,82). Regulatory networks involving long (l)ncRNAs further augment this layer of control. For example, the lncRNA KCNQ1OT1 serves as a competitive endogenous RNA to sponge miR-192-5p, thereby upregulating the anti-apoptotic protein X-linked inhibitor of apoptosis (83), while the lncRNA ZFAS1 protects against apoptosis by sequestering miR-34b-5p to upregulate sirtuin (SIRT)1 (84). Conversely, detrimental miRNAs such as miR-208a-5p and miR-21-3p promote apoptosis (85).

Additionally, miRNAs participate in specialized cell death modalities and mitochondrial dysfunction associated with SIMD. miR-383-3p has been shown to alleviate SIMD by inhibiting ferroptosis via the activating transcription factor 4 (ATF)-CHOP-ChaC glutathione specific γ-glutamylcyclotransferase 1) (86), whereas miR-194-5p aggravates oxidative stress and apoptosis by targeting DUSP9 (87). The Xist/miR-7a-5p pathway serves a key role in sepsis-induced mitochondrial dysfunction. Inhibition of either Xist or miR-7a-5p upregulates the key mitochondrial biogenesis factor PGC-1α, increases ATP production, and decreases cardiomyocyte apoptosis, highlighting their key role in maintaining mitochondrial homeostasis (88,89).

Natural products against SIMD

The pathophysiology of SIMD is complex and multifactorial. Interplay of cytokine storm, excessive generation of ROS and RNS, mitochondrial dysfunction and dysregulated endothelial-myocardial crosstalk collectively drive cardiomyocyte apoptosis, ferroptosis and microcirculatory impairment.

Regulation of programmed cell death

Programmed cell death pathways implicated in SIMD include apoptosis, pyroptosis, autophagy and ferroptosis. Natural products can confer cardioprotection by either enhancing cell survival signals or inhibiting specific death pathways (Fig. 2).

Regulation of programmed cell death
by natural products in SIMD. Natural products attenuate SIMD
through suppression of pro-oxidant NF-κB/TLR signaling, modulation
of PI3K/Akt, AMPK/autophagy, TFEB, NLRP3/pyroptosis and mitophagy
pathways and restoration of GPX4-associated antioxidant defenses.
SIMD, sepsis-induced myocardial dysfunction; TLR, Toll like
receptor; TFEB, T cell transcription factor EB; GPX, Glutathione
peroxidase; SphK, Sphingosine kinases; S1P, Sphingosine Kinase 1;
ROS, reactive oxygen species; MFN, mitofusin; SIRT, sirtuin 1;
GSDMD, gasdermin D; ALOX, arachidonate 5-lipoxygenase.

Figure 2

Regulation of programmed cell death by natural products in SIMD. Natural products attenuate SIMD through suppression of pro-oxidant NF-κB/TLR signaling, modulation of PI3K/Akt, AMPK/autophagy, TFEB, NLRP3/pyroptosis and mitophagy pathways and restoration of GPX4-associated antioxidant defenses. SIMD, sepsis-induced myocardial dysfunction; TLR, Toll like receptor; TFEB, T cell transcription factor EB; GPX, Glutathione peroxidase; SphK, Sphingosine kinases; S1P, Sphingosine Kinase 1; ROS, reactive oxygen species; MFN, mitofusin; SIRT, sirtuin 1; GSDMD, gasdermin D; ALOX, arachidonate 5-lipoxygenase.

Inhibition of apoptosis

Cardiomyocyte apoptosis is an early pathogenic event in SIMD, contributing to amplified inflammation and progressive cardiac dysfunction.

NF-κB, a master regulator of inflammation and apoptosis, is a common target for numerous natural compounds that also inhibit upstream TLR signaling. In CLP-induced septic mice, compounds such as gramine, curcumin, andrographolide and berberine suppress the TLR1/NF-κB axis, leading to lower TNF-α, IL-1β and NO levels, decreased caspase-3 activation and apoptosis and restored cardiac contractility, which translates into decreased mortality (90-94). Notoginsenoside R1 activates the PI3K/Akt pathway while concurrently inhibiting NF-κB, resulting in suppressed caspase-3 activation and downregulated TNF-α and IL-1β levels in both cardiomyocyte H9c2 and septic mice, thereby attenuating inflammation and apoptosis (95,96).

The MAPK family also influences stress-induced apoptosis. In LPS-challenged septic mice, tetrahydrocurcumin upregulates mitogen activated protein kinase phosphatase 1), suppresses ERK and JNK signaling and limits ROS production and caspase-3-mediated apoptosis, preserving cardiac function (97). Astaxanthin similarly inhibits ERK, JNK and p38 MAPK pathways while activating PI3K/Akt pathway, resulting in lower serum levels of TNF-α, IL-6 and BNP, and reduced myocardial injury and apoptosis in septic mice (98). Furthermore, 1-deoxynojirimycin decreases ROS generation by inhibiting JAK2/STAT6 signaling, thereby decreasing cardiomyocyte apoptosis and improving function in septic mice (99). Astragaloside IV protects against LPS-induced myocardial injury in rats by inhibiting the JNK/Bax pathway, reducing levels of caspase-3, CK-MB (Creatine kinase (CK-MB) and c-TnI (Cardiac troponin I c-TnI) and increasing the Bcl-2/Bax ratio to mitigate myocardial dysfunction (100).

In CLP-induced septic rats, apigenin inhibits the Sphingosine kinase 1/sphingosine 1-phosphate) pathway, leading to downregulation of cleaved caspase-3/-9 and Bax, upregulation of Bcl-2, decreased serum CK-MB and lactate dehydrogenase (LDH) levels, suppressed apoptosis and improved myocardial histology (101). Gastrodin, tested in CLP-induced septic mice and AC16 cardiomyocytes, modulates the denticleless E3 ubiquitin protein ligase-homolog histone acetyltransferase-driven ubiquitination-acetylation axis to promote degradation of pro-caspase-3/-9 and Bax, thereby attenuating apoptosis and alleviating myocardial damage (102).

Inhibition of pyroptosis

Pyroptosis is an inflammatory form of programmed cell death triggered by inflammasome activation, characterized by GSDMD-mediated pore formation and the release of IL-1β and IL-18 (103). In SIMD, pyroptosis not only accelerates cardiomyocyte loss but also perpetuates a local inflammatory cycle.

In CLP-induced septic mice, vaccarin and ruscogenin attenuate myocardial injury by suppressing the NLRP3 inflammasome, decreasing IL-1β and IL-18 release and limiting pyroptosis (103,104). Artemisinin decreases NLRP3 and caspase-1 expression in burned septic mice, lowers IL-1β and IL-18 mRNA in mouse monocytic macrophage leukemia RAW264.7 cells and decreases neutrophil infiltration, thereby mitigating myocardial damage (105). Similarly, thymoquinone inhibits the NLRP3/caspase-1/IL-1β/IL-18 pathway, downregulates p62 and c-TnT and upregulates IL-10, reducing inflammation and pyroptosis in septic mice (106). Additionally, in LPS-challenged septic mice, syringaresinol activates the endoplasmic reticulum (ER)/SIRT1 pathway and suppresses NLRP3/GSDMD signaling to block IL-1β and IL-18 release, significantly improving myocardial injury (107). Both ginsenoside Rg1 and gastrodin inhibit the TLR4/NF-κB/NLRP3 signaling cascade, downregulate Bax, upregulate Bcl-2 and restore cardiac function by reducing pyroptosis (108,109). Nifuroxazide similarly targets TLR4/NLRP3/IL-1β signaling to lower LDH and CK-MB levels, suppress pyroptosis and protect myocardial function (110).

ROS also regulate NLRP3 activation. In LPS-induced septic mice, carvacrol and emodin inhibit the ROS/NLRP3/GSDMD pathway, decreasing IL-1β and IL-18 levels, which enhances survival rate and restores cardiac function (111,112). Plumbagin also suppresses ROS-mediated NLRP3/GSDMD signaling, decreases HMGB1, caspase-3 and Bax expression and improves cardiac function through preventing pyroptosis (113). Geniposide downregulates neutrophil cytoplasmic factor 1 and suppresses the AMPKα/ROS/NLRP3 pathway, leading to reduced ROS levels and improved cardiac function and survival rate in septic mice (114). Finally, oxycodone markedly improves EF and fractional shortening (FS), decreases levels of ROS, c-TnI and CK-MB and activates the Nrf2/heme oxygenase (HO)-1 pathway to inhibit NLRP3-mediated pyroptosis, yielding notable cardioprotective effects (115).

Autophagy

Autophagy is a key homeostatic process that removes damaged organelles and proteins via lysosomal degradation. In cardiomyocytes, it maintains mitochondrial quality, balances energy metabolism and supports antioxidant defenses (39). In SIMD, autophagy exhibits a dual role: Moderate activation clears dysfunctional mitochondria and limits oxidative stress, whereas either excessive or insufficient autophagic flux can exacerbate cell death and impair cardiac function (116).

The AMP-activated protein kinase (AMPK) pathway induces autophagy. In CLP-induced septic mice, luteolin activates AMPK pathway signaling, which increases LC3-II and Beclin-1 expression, enhances autophagic flux, improves LVEF and FS, decreases levels of TNF-α, IL-6 and ROS, restores mitochondrial architecture and inhibits apoptosis (116). Conversely, in CLP-induced septic rats, tangeretin suppresses excessive autophagy by inhibiting PTEN and activating the Akt/mTOR pathway. This lowers serum levels of cardiac myosin light chain-1 (cMLC1), c-TnI, LDH and (CK), alleviates oxidative stress and inflammation and preserves myocardial function (117).

The NF-κB/TFEB (T cell transcription factor EBTFEB pathway also regulates autophagy. In LPS-challenged septic mice, apigenin inhibits NF-κB to decrease inflammation and oxidative stress while upregulating TFEB and its downstream target genes such as ATG5, lysosome-associated membrane protein 1), p62, microtubule-associated protein 1 light chain 3), vacuolar protein sorting 11), thereby boosting autophagic flux. These combined effects lower cardiac injury markers (CK, LDH, c-TnI, cMLC1), improve survival rate and maintain cardiac function (118,119).

Mitochondrial autophagy (mitophagy) is key for clearing damaged mitochondria. In LPS-stimulated H9c2 cardiomyocytes, puerarin upregulates Drp1 and mitofusin 1 to preserve mitochondrial dynamics and activates the PINK1/Parkin pathway, thereby reducing ROS, inhibiting apoptosis and restoring cell viability (120).

Other natural compounds modulate autophagy through alternative signaling nodes. In LPS-induced septic mice, narciclasine and polydatin activate SIRT6 and JNK signaling, which elevates LC3-II and Beclin-1 while lowering TNF-α and IL-6 levels, thereby suppressing inflammation and attenuating myocardial injury (121,122). Phlorizin modulates the Hif-1α/BCL2 interacting protein 3) axis to promote Beclin-1 release, autophagosome formation and lysosomal degradation, reducing oxidative stress and apoptosis, improving cardiac function and enhancing survival rate (123).

Inhibition of ferroptosis

Ferroptosis is an iron-dependent form of programmed cell death characterized by glutathione peroxidase 4 (GPX4) inactivation, accumulation of lipid peroxides and iron overload (37). In SIMD, inflammatory disruption of iron homeostasis coupled with mitochondrial dysfunction accelerates lipid peroxidation, triggering ferroptosis and exacerbating cardiomyocyte injury (124). Consequently, targeting antioxidant defenses, iron metabolism and lipid peroxidation represents a promising therapeutic strategy.

The AMPK signaling axis serves a central role in regulating ferroptosis. In LPS-induced septic mice, puerarin activates AMPK to upregulate GPX4 and ferritin and downregulate ACSL4 and the transferrin receptor (TFRC), reducing myocardial iron content, lipid peroxidation and ferroptosis (124). Curcumin engages the AMPK/SIRT1 pathway to increase expression of GPX4, ferritin and translocase of outer mitochondrial membrane 20 while decreasing levels of Fe2+, MDA and prostaglandin-endoperoxide synthase 2, thereby preventing ferroptotic cell death (125). Similarly, matrine activates PI3K/Akt to enhance GPX4 and the Bcl-2/Bax ratio, reduce ACSL4 and ROS and inhibit ferroptosis, improving cardiac performance (126). Ginsenoside Rg1 stimulates FAK/Akt and upregulates FOXO3A, which decreases levels of TNF-α, IL-1β and Fe2+ and increases Bcl-2, collectively alleviating myocardial injury (127).

Direct inhibition of key lipid peroxidation enzymes provides another mechanism to block ferroptosis. In LPS-stimulated AC16 cardiomyocytes, resveratrol upregulates miR-149 and downregulates HMGB1, decreasing Fe2+ accumulation and lipid ROS to protect the myocardium (128). In CLP-induced septic mice, narciclasine restores glutathione, downregulates TFRC and upregulates GPX4 and HO-1, thereby inhibiting lipid peroxidation and preserving cardiac function (129). Furthermore, resveratrol activates the SIRT1/Nrf2 pathway to mitigate mitochondrial damage and lipid peroxidation (130). Arachidonic acid 15-lipoxygenase (ALOX15) is a key enzyme in lipid peroxidation. Wogonin directly inhibits ALOX15, mitigating lipid peroxidation and ferroptosis in the myocardium (131). Quercetin also activates PI3K/Akt to inhibit ALOX5-mediated lipid oxidation, restores GPX4 and glutathione (GSH) levels, lowers Fe2+ and ROS, and suppresses myocardial inflammation and ferroptosis (132).

Collectively, natural products, such as flavonoids, alkaloids and saponins, attenuate SIMD through diverse mechanisms. These include suppression of pro-oxidant NF-κB/TLR signaling, modulation of PI3K/Akt, AMPK/autophagy, TFEB, NLRP3/pyroptosis and mitophagy pathways and restoration of GPX4-related antioxidant defenses. For example, curcumin, resveratrol, and quercetin concurrently inhibit apoptosis and pyroptosis, promote adaptive autophagy and restrain ferroptosis. Similarly, astaxanthin, apigenin and puerarin protect the myocardium through combined activation of autophagy and inhibition of ferroptosis. This multimodal pharmacological profile highlights the potential of natural products to target the interconnected cell death networks that drive SIMD.

Regulation of inflammation

Sepsis provokes a systemic inflammatory response that contributes directly to myocardial injury. Endotoxins and other PAMPs activate key signaling cascades, most notably the TLR4/NF-κB, MAPK and NLRP3 inflammasome pathways, leading to the release of proinflammatory cytokines (TNF-α, IL-1β, IL-6, MCP-1) and recruitment of neutrophils and macrophages into cardiac tissue. The resulting inflammatory milieu impairs cardiomyocyte contractility and survival (Fig. 3).

Anti-inflammatory mechanisms of
natural products in sepsis-induced myocardial dysfunction). Natural
products confer protection by inhibiting pro-inflammatory
TLR4/NF-κB and MAPK signaling cascades, activating the PI3K/Akt
survival pathway, modulating specific miR networks (miR-214-5p,
miR-21) and promoting the polarization of macrophages towards the
reparative M2 phenotype. TLR, toll-like receptor; miR, miRNA; GSK,
glycogen synthase kinase; HO, heme oxygenase; SIRT, sirtuin 1; COX,
cyclooxygenase; iNOS, inducible nitric oxide synthase; IGF, insulin
like growth factor; LDH, lactate dehydrogenase; MCP, monocyte
chemotactic protein; MDA, malondialdehyde; HMGB, high mobility
group box.

Figure 3

Anti-inflammatory mechanisms of natural products in sepsis-induced myocardial dysfunction). Natural products confer protection by inhibiting pro-inflammatory TLR4/NF-κB and MAPK signaling cascades, activating the PI3K/Akt survival pathway, modulating specific miR networks (miR-214-5p, miR-21) and promoting the polarization of macrophages towards the reparative M2 phenotype. TLR, toll-like receptor; miR, miRNA; GSK, glycogen synthase kinase; HO, heme oxygenase; SIRT, sirtuin 1; COX, cyclooxygenase; iNOS, inducible nitric oxide synthase; IGF, insulin like growth factor; LDH, lactate dehydrogenase; MCP, monocyte chemotactic protein; MDA, malondialdehyde; HMGB, high mobility group box.

Inhibition of the TLR4/NF-κB pathway

Numerous flavonoids and other phytochemicals suppress TLR4-driven NF-κB activation to decrease cytokine release and improve cardiac function. In LPS-challenged septic mice, isoquercitrin and quercetin inhibit NF-κB p65 phosphorylation, lower TNF-α, IL-6 and MCP-1 levels and significantly improve LVEF and FS (133,134). Ciprofol similarly blocks NF-κB activation in septic mice, decreasing levels of CK-MB, LDH, TNF-α and IL-6 and restoring LVEF and FS (135). In CLP-induced septic rats, astragaloside IV prevents NF-κB p65 phosphorylation, decreases levels of LDH, CK-MB, IL-6, IL-1β and HMGB1 and enhances both systolic and diastolic function as well as survival rate (136). Myricetin inhibits IκBα degradation and NF-κB/p65 nuclear translocation in septic rats, which lowers TNF-α, IL-6 and IL-1β and preserves cardiac function (137,138). Similarly, naringin blocks NF-κB nuclear translocation, decreases TNF-α, IL-1β and IL-6 levels and enhanced superoxide dismutase activity while lowering MDA levels and leads to improved cardiac performance in septic rats (139,140). Additionally, astilbin and esculin both target the TLR4/NF-κB pathway to downregulate TNF-α, IL-6 and MDA, correct QT prolongation and reduce the risk of ventricular arrhythmia (141,142). Puerarin also inhibits TLR4/NF-κB signaling in Langendorff-perfused hearts, decreasing LDH, CK and TNF-α release and improving LV end-diastolic pressure in septic rats (143). By contrast, oleuropein activates the glycogen synthase kinase (GSK)-3 beta)/NF-κB pathway, decreases c-TnI, CK-MB, IL-6 and HMGB1 levels and increases anti-inflammatory IL-10 levels, thereby protecting cardiac function in septic rats (144).

Inhibition of MAPK-associated pathways

The MAPK family intersects with NF-κB signaling to amplify inflammation. In LPS-stimulated H9C2 cells, zerumin A suppresses MAPK-mediated NF-κB activation, resulting in lower levels of iNOS, ROS, COX-2, NO, MCP-1, TNF-α, IFN-γ and IL-1β and higher IL-10 expression, which improves cell viability (145). In LPS-challenged septic mice, propofol and carvacrol both inhibit ERK1/2 phosphorylation, decrease IL-6 and TNF-α levels, alleviate myocardial injury and enhance survival rate (146,147). Pinocembrin blocks the p38/JNK MAPK pathway, decreasing myocarditis and arrhythmia risk in LPS-induced septic rats (148). Similarly, madecassoside inhibits ERK1/2 and p38 activation in septic mice, delays arterial pressure decline and lowers TNF-α levels, thereby mitigating tachycardia (149). Additionally, alisol B 23-acetate suppresses TLR4/NOX2 and p38/MAPK/ERK signaling, decreasing IL-6, IL-1β and TNF-α levels, which alleviates myocardial injury and improves survival rate (150). In LPS-treated septic rats, oxymatrine also inhibits the p38/MAPK/caspase-3 and JAK2/STAT3 pathways, decreases IL-1β and TNF-α levels and improves cardiac function (151,152).

Activation of the PI3K/AKT pathway

The PI3K/Akt pathway exerts anti-inflammatory and pro-survival effects in SIMD. In LPS-challenged septic mice, astragaloside IV activates PI3K/Akt signaling to reduce LDH and c-TnI levels, suppress myocardial inflammation and improve cardiac performance (153). Paeoniflorin and resveratrol also stimulate PI3K/Akt to decrease levels of TNF-α, IL-1β, IL-6, IL-12, MCP-1, IFN-γ and iNOS, thereby protecting against myocardial injury (154,155). In CLP-induced septic mice, thaliporphine activates the PI3K/Akt/mTOR pathway to decrease levels of TNF-α, c-TnI and LDH and enhance LV function (156). Salidroside modulates the IGF-1/PI3K/Akt/GSK-3β pathway to decrease levels of CK, LDH, TNF-α, IL-6 and IL-1β, thereby alleviating myocardial edema and dysfunction (157).

miRNA-dependent anti-inflammatory pathways

miRNAs have emerged as critical regulators of myocardial inflammation in sepsis. For example, corylin improves LPS-induced cardiac dysfunction in mice by downregulating myocardial miR-214-5p, which attenuates inflammatory signaling (158). In CLP-induced septic mice, hyperoside exerts protective effects by suppressing the sepsis-associated upregulation of miR-21, leading to decreased myocardial inflammation (159). Furthermore, matrine acts via the PTENP1/miR-106b-5p pathway to inhibit proinflammatory pathways and enhance cardiomyocyte viability, thereby improving cardiac function in CLP-induced mice (160). Chrysophanol downregulates miR-27b-3p and upregulates PPARγ, which suppresses inflammatory cytokines and apoptotic proteins to repair CLP-induced cardiac injury in mice (161).

Targeting macrophage polarization

Macrophage polarization strongly influences the inflammatory milieu in SIMD. Pinostrobin inhibits TLR4/myeloid differentiation protein-2 signaling in mouse monocytic macrophage leukemia RAW264.7cells, decreasing expression of NO, prostaglandin E2, TNF-α, IL-12, iNOS and COX-2, and improves heart rate and survival rate in LPS-treated zebrafish (162). Cynaroside activates the Nrf2/HO-1 pathway to lower IL-1β and TNF-α levels and promotes M2 macrophage polarization, thereby decreasing systemic inflammation and protecting the myocardium in septic mice (163). Similarly, ginsenoside Rc suppresses the STAT3/FoxO3a/Sirt1 pathway to limit M1 macrophage-mediated myocardial damage during sepsis (164).

Other anti-inflammatory agents

Several additional natural products exhibit potent anti-inflammatory and cardioprotective effects in SIMD. In LPS-induced septic mice, cyanidin lowers levels of LDH, CK, c-TnI, TNF-α, IL-1β, MIP-2, MCP-1 and cMLC1, inhibits caspase-3 and PARP cleavage and attenuates myocardial injury (165). Monotropein and baicalein suppress TNF-α, IL-6, HMGB1 and iNOS/NO production while decreasing MMP-2/9 and ROS levels to alleviate septic myocardial hypertrophy and dysfunction (166,167). Paeoniflorin and honokiol markedly decrease c-TnI, LDH, IL-6, TNF-α and IL-1β and raise IL-10 levels, improving cardiac contractility and enhancing survival rate in CLP-induced septic mice (168,169). Similarly, dobutamine increases serum levels of IL-10 and lowers c-TnI, NT-proBNP and H-FABP, thereby enhancing survival rate in CLP-induced rats (170).

In LPS-challenged septic rats, magnolol corrects hypotension and bradycardia by decreasing levels of iNOS, NO, TNF-α and alanine aminotransferase/aspartate aminotransferase) (171). Sodium tanshinone IIA sulfonate also improves LV function by inhibiting expression of TNF-α, IL-6, HMGB1, C-reactive protein, PCT (Procalcitonin (PCT), c-TnI/c-TnT and BNP (172). Additionally, micromeria congesta downregulates IL-2, caspase-3 and heat shock protein (HSP)-27 to attenuate septic myocardial pathology (173). Naringenin and tubeimoside I activate the SIRT3 or HIF-1α pathways to suppress IL-1β, IL-6 and TNF-α, improving myocardial injury (174,175). Yohimbine blocks α2A-adrenergic receptors at cardiac sympathetic terminals, promotes norepinephrine release and decreases NO and TNF-α levels to alleviate cardiac dysfunction in septic rats (176).

In summary, these natural products protect against SIMD by inhibiting TLR4/NF-κB and MAPK signaling, activating PI3K/Akt, modulating miRNA networks and promoting macrophage M2 polarization. They collectively reduce proinflammatory cytokine levels, limit inflammatory cell infiltration and improve cardiac function and survival rate.

Regulation of oxidative stress

Oxidative stress is a key driver of cardiac injury in SIMD. Inflammation, endotoxins and mitochondrial dysfunction increase ROS/RNS levels, leading to lipid peroxidation, protein carbonylation and DNA damage (49). These changes worsen contractile dysfunction and promote cardiomyocyte death (Fig. 4).

Modulation of oxidative stress by
natural products in sepsis-induced myocardial dysfunction). Natural
products primarily enhance antioxidant defenses by activating
enzymes such as SOD, CAT and GSH-Px or by stimulating the Nrf2/HO-1
pathway. They also dampen proinflammatory signaling via NF-κB, TLR4
and ERK1/2-p38 MAPK signaling to reduce NOX2/iNOS-mediated ROS/RNS
production. SOD, superoxide dismutase; CAT, catalase; GSH-Px,
glutathione peroxidase; HO, heme oxygenase; TLR, Toll like
receptor; NOX, nitrogen oxide; iNOS, inducible nitric oxide
synthase; ROS, reactive oxygen species; RNS, reactive nitrogen
species; GSSG, glutathione oxidized; MDA, malondialdehyde; LDH,
lactate dehydrogenase; TAK, transforming growth factor-β-activated
kinase 1.

Figure 4

Modulation of oxidative stress by natural products in sepsis-induced myocardial dysfunction). Natural products primarily enhance antioxidant defenses by activating enzymes such as SOD, CAT and GSH-Px or by stimulating the Nrf2/HO-1 pathway. They also dampen proinflammatory signaling via NF-κB, TLR4 and ERK1/2-p38 MAPK signaling to reduce NOX2/iNOS-mediated ROS/RNS production. SOD, superoxide dismutase; CAT, catalase; GSH-Px, glutathione peroxidase; HO, heme oxygenase; TLR, Toll like receptor; NOX, nitrogen oxide; iNOS, inducible nitric oxide synthase; ROS, reactive oxygen species; RNS, reactive nitrogen species; GSSG, glutathione oxidized; MDA, malondialdehyde; LDH, lactate dehydrogenase; TAK, transforming growth factor-β-activated kinase 1.

Activation of antioxidant enzymes

Natural products directly enhance the activity of endogenous antioxidant systems. In LPS-induced septic mice, lycopene, echinacoside and apocynin lower myocardial ROS and MDA levels while increasing NADPH oxidase and SOD activity (177-179). Similarly, rutin reduces CK, LDH, MDA, ROS, TNF-α and IL-6 and increases SOD and catalase (CAT) activity, thereby protecting cardiac structure and function (180). In CLP-induced rats, curcumin decreases plasma levels of ROS, c-TnI and MDA, restores the GSH/oxidized glutathione (GSSG) ratio and enhances SOD activity to improve LVEF and FS (181,182). Quercetin similarly upregulates endothelial (e)NOS and maintains the GSH/GSSG balance, alleviating hypotension and tachycardia in septic mice (183).

Activation of the Nrf2 pathway

The transcription factor Nrf2 orchestrates the cellular antioxidant response. In LPS-challenged septic mice, resveratrol activates Nrf2 to decrease myocardial ROS and increase the expression of antioxidant genes (184). Chrysin and cardamonin engage the ERK-Nrf2-HO-1 axis, upregulating HO-1 and SOD while lowering levels of ROS, MDA, TNF-α, IL-6 and IL-1β, which improves cardiac function (185,186). Dehydrocorydaline also triggers the Nrf2/HO-1 pathway, decreases levels of ROS, TNF-α and IL-1β and restores SOD and glutathione peroxidase (GSH-Px) activity, leading to improved survival rate (187).

Regulation of the PI3K/Akt/NF-κB pathway

NF-κB links inflammation with oxidative stress. In LPS-treated septic rats, baicalein inhibits the NF-κB pathway to decrease levels of iNOS, ROS and NO, improving blood pressure, heart rate and survival rate (188,189). Thaliporphine also suppresses the TLR4/transforming growth factor-β-activated kinase 1 (TAK1)/NF-κB pathway to lower myocardial NO, ROS, TNF-α and iNOS levels and improve LV pressure-volume indices (190). Ginsenoside Re corrects the iNOS/eNOS imbalance by inhibiting NF-κB signaling, thereby reversing myocardial injury (191). In LPS-treated rats, salidroside activates the PI3K/Akt/mTOR pathway while inhibiting NF-κB, which decreases levels of iNOS, COX-2 and ROS and improves cardiac function (192). Propofol also engages mTOR signaling to lower ROS and MDA levels and boosts SOD activity, thereby reverse myocardial damage (193).

Other antioxidant agents

Additional natural products modulate oxidative stress via unique pathways. In LPS-induced sepsis, ginkgolide A activates FoxO1 and downstream effectors such as Kruppel-like factor 15, thioredoxin 2), Notch1(Notch receptor 1), XBP1(X-box binding protein 1), which upregulates antioxidant enzymes and preserves mitochondrial function (194). Tanshinone IIA inhibits ERK1/2 and p38 MAPK phosphorylation, decreases NOX2 expression and lowers MDA and ROS levels to alleviate cardiac dysfunction (195).

In summary, natural products primarily enhance antioxidant defenses by activating enzymes such as SOD, CAT and GSH-Px or by stimulating the Nrf2/HO-1 pathway. They also dampen proinflammatory signaling via NF-κB, TLR4 and ERK1/2/p38 MAPK pathways to reduce NOX2/iNOS-mediated ROS/RNS production. Through simultaneous engagement of PI3K/Akt/mTOR and FoxO1 pathways, these compounds preserve mitochondrial integrity and myocardial contractility, thereby markedly improving SIMD outcomes and survival rate.

Regulation of mitochondrial dysfunction

Mitochondria serve not only as the energy source of cardiomyocytes but also as a key hub for ROS production and the regulation of programmed cell death (54). In SIMD, inflammatory mediators, oxidative stress and impaired mitochondrial quality control cause mitochondrial fragmentation, loss of membrane potential, respiratory defects, diminished ATP synthesis and ultimately cell death (55) (Fig. 5).

Targeting mitochondrial dysfunction
with natural products in SIMD. Natural products counteract SIMD by
engaging multiple mitochondrial-protective pathways, most notably
SIRT1/Nrf2, PI3K/Akt/mTOR and AMPK, and by promoting mitochondrial
biogenesis via PGC-1α/TFAM and UPRmt. SIMD, sepsis-induced
myocardial dysfunction; SIRT, sirtuin 1; PGC, peroxisome
proliferator-activated receptor γ coactivator; UPRmt, mitochondrial
unfolded protein response; GSK, glycogen synthase kinase; PTP1B,
protein tyrosine phosphatase 1B; ROS, reactive oxygen species; SOD,
superoxide dismutase; DRP, dynamin-related protein 1; STIM, stromal
interaction molecule 1; CRAC, calcium release-activated
channels.

Figure 5

Targeting mitochondrial dysfunction with natural products in SIMD. Natural products counteract SIMD by engaging multiple mitochondrial-protective pathways, most notably SIRT1/Nrf2, PI3K/Akt/mTOR and AMPK, and by promoting mitochondrial biogenesis via PGC-1α/TFAM and UPRmt. SIMD, sepsis-induced myocardial dysfunction; SIRT, sirtuin 1; PGC, peroxisome proliferator-activated receptor γ coactivator; UPRmt, mitochondrial unfolded protein response; GSK, glycogen synthase kinase; PTP1B, protein tyrosine phosphatase 1B; ROS, reactive oxygen species; SOD, superoxide dismutase; DRP, dynamin-related protein 1; STIM, stromal interaction molecule 1; CRAC, calcium release-activated channels.

Activation of Nrf-associated pathways

Nrf pathways are key for maintaining mitochondrial redox homeostasis. In LPS-induced septic mice, curcumin and resveratrol upregulate PGC-1α and mitochondrial transcription factor A (TFAM), inhibit DRP1-mediated excessive mitochondrial fission and activate the SIRT1/Nrf2 pathway to normalize mitochondrial morphology and respiration, thereby markedly improving cardiac function (196,197). Rosmarinic acid similarly preserves mitochondrial membrane potential and ATP production by triggering the SIRT1/Nrf2 pathway, resulting in decreased ROS levels and enhanced survival rate (198). Songorine further boosts Nrf1/TFAM signaling and promotes PGC-1α-Nrf2 interactions, suppressing calcium release-activated channels) channel formation, lowering mitochondrial ROS levels, restoring calcium homeostasis and increasing contractile power (199).

Inhibition of AMPK-associated pathways

Although AMPK often supports mitochondrial homeostasis, certain stress contexts benefit from modulating its downstream effectors. In LPS-stimulated H9C2 cells, hesperetin upregulates Bcl-2, downregulates Bax and caspase-3/9 expression through inhibiting JNK phosphorylation, thus preserving mitochondrial membrane potential and decreasing apoptosis (200). In CLP-induced septic mice, tannic acid attenuates ER stress and decreases expression of ROS, Bax, cytochrome c and caspase-3/-9/-12, thereby safeguarding mitochondrial function in cardiomyocytes (201). Additionally, astaxanthin inhibits the PTP1B/JNK pathway to maintain membrane potential and limit ROS generation, which decreases cardiomyocyte apoptosis (202). In LPS-challenged septic mice, lycorine and naringenin activate AMPK and suppress NF-κB signaling, together protecting cardiac function (203,204). Obeticholic acid blocks the ERK1/2-DRP1 axis to prevent cytochrome c release and upregulates GPX1 and SOD1/2, thereby restoring mitochondrial integrity (205).

Activation of PI3K/Akt signaling

The PI3K/Akt pathway also contributes to mitochondrial preservation. In LPS-induced septic rats, neferine activates PI3K/Akt/mTOR signaling to elevate the Bcl-2/caspase-3 ratio, decreases ROS levels and restores mitochondrial structure and function, leading to improved myocardial performance (206). Ginsenoside Rg1 also engages the recombinant purinergic receptor P2X to stimulate the Akt/GSK-3β pathway, suppressing mitochondrial superoxide generation and cytochrome c release, stabilizing calcium handling and enhancing survival rate (207).

Other pathways

In LPS-treated septic mice, verbascoside promotes mitochondrial biogenesis and morphological repair, thereby improving cardiac function (208). Silibinin also decreases CCR2 levels and activates LXRα (Liver X receptors) signaling to dampen inflammation and oxidative damage while improving mitochondrial function (209,210). Additionally, chicoric acid inhibits α-tubulin acetylation and NLRP3 inflammasome assembly, indirectly restoring mitochondrial architecture and alleviating myocardial injury (211). Songorine also activates Wnt/β-catenin signaling to boost mitochondrial biogenesis and suppress inflammation and apoptosis, thereby improving LVEF and FS (212). Furthermore, excavatolide B modulates intracellular and extracellular Ca2+ channels to reverse calcium dysregulation and repair mitochondrial structure in septic hearts (213). Salvianolic acid B induces an ATF5-mediated mitochondrial unfolded protein response (UPRmt), prevents excessive mitochondrial fission, preserves membrane potential and significantly enhances contractile function and mitochondrial ultrastructure (214).

In summary, natural products counteract SIMD by engaging multiple mitochondrial-protective pathways, most notably SIRT1/Nrf2, PI3K/Akt/mTOR and AMPK pathways, and by promoting mitochondrial biogenesis via PGC-1α/TFAM and UPRmt. These interventions preserve mitochondrial membrane potential, augment ATP production, decrease ROS accumulation and suppress apoptosis, collectively leading to substantial improvements in cardiac function and survival rate in SIMD.

Regulation of endothelial-myocardial crosstalk

ECs constitute the first line of defense in maintaining microcirculatory stability and myocardial function (61). In sepsis, endothelial barrier dysfunction increases vascular permeability, tissue edema and inflammatory cell infiltration, while endothelial-derived cytokines and exosomes propagate injurious signals to cardiomyocytes, exacerbating SIMD (63).

In LPS-induced septic mice, neohesperidin dihydrochalcone markedly suppresses vascular hyperpermeability and myocardial tissue damage. It decreases ROS production, preserves mitochondrial membrane potential and enhances antioxidant defenses (CAT, SOD, GSH) while lowering MDA levels (215). Mechanistically, it inhibits phosphorylation of TAK1, ERK1/2 and NF-κB, stabilizes endothelial junction protein and diminishes THP-1 monocyte adhesion and infiltration (215). These combined effects reinforce endothelial barrier function and mitigate myocardial injury (215). In LPS-induced septic shock rats, anisodamine restores hemodynamics and repairs the endothelial glycocalyx, thereby decreasing myocardial damage (8). It lowers levels of lactate, IL-1β, IL-6, TNF-α, CK, c-TnT, NT-proBNP, syndecan-1 and hyaluronic acid via inhibition of NF-κB and NLRP3 signaling and activation of the PI3K/Akt pathway (8). Moreover, exosomes released by LPS-stimulated human umbilical vein ECs transfer inflammatory and oxidative signals to the myocardium; anisodamine attenuates these exosome-mediated effects, indicating that it disrupts harmful endothelial-myocardial crosstalk to confer cardioprotection in SIMD (216).

Classification and structure-activity relationships

Natural products with efficacy against SIMD can be divided into five major chemical classes: Flavonoids, glycosides, phenolic acids, alkaloids and saponins (Fig. 6A). Notably, flavonoids and glycosides account for approximately half of the identified compounds. The prominent bioactivity of these classes is associated with their characteristic chemical scaffolds. Specifically, the presence of multiple hydroxyl (-OH) groups confers potent antioxidant capacity by serving as hydrogen donors to directly neutralize free radicals (217). Concurrently, conjugated double bond systems (extended π-electron networks) enable electron delocalization, which stabilizes antioxidant reaction intermediates and facilitates key interactions with cellular signaling proteins (kinases, transcription factors) (217). These combined physicochemical properties underpin their broad-spectrum antioxidant, anti-inflammatory and cytoprotective activities observed in SIMD models (Fig. 6B).

Classification and structure-activity
analysis of natural products active against sepsis-induced
myocardial dysfunction). (A) Active natural products are mainly
divided into flavonoids, glycosides, phenolic acids, alkaloids and
saponins, lignin and olefins. (B) Pharmacological activity analysis
of natural products shows that functional groups are associated
their antioxidant, anti-inflammatory and cell protective
activities.

Figure 6

Classification and structure-activity analysis of natural products active against sepsis-induced myocardial dysfunction). (A) Active natural products are mainly divided into flavonoids, glycosides, phenolic acids, alkaloids and saponins, lignin and olefins. (B) Pharmacological activity analysis of natural products shows that functional groups are associated their antioxidant, anti-inflammatory and cell protective activities.

Flavonoids

Flavonoids share a characteristic C6-C3-C6 three-ring scaffold. Critical structural features include a C2-C3 double bond, hydroxyl groups at C3, C5 and/or C7 and a catechol-type dihydroxy arrangement on the B-ring (positions 3',4') (217). This configuration maximizes electron delocalization and hydrogen-donating capacity, explaining their strong radical scavenging activity and their direct, Structure-activity relationship (SAR)-driven inhibition of central pro-inflammatory signaling hubs such as NF-κB and MAPK (218). Mechanistically, flavonoids thereby modulate the TLR4/NF-κB pathway, NLRP3 inflammasome activation and MAPK pathways to lower proinflammatory cytokines (IL-1β, TNF-α) and attenuate cardiomyocyte apoptosis (219).

Glycosides

Glycosides consist of aglycone cores (flavonoid, sesquiterpene or other scaffolds) linked to sugar moieties via O- or C-glycosidic bonds. Glycosylation is a key SAR-modifying factor: It markedly enhances aqueous solubility and pharmacokinetic properties, improving tissue distribution and bioavailability (220). Notable glycosides in SIMD include isoquercitrin, paeoniflorin and salidroside. The sugar units promote interactions with cell-surface receptors or transporters, facilitate macrophage M2 polarization and improve cardiomyocyte survival rate (220). Glycosides often retain and enhance the anti-inflammatory and anti-apoptotic properties of their parent aglycones, while also demonstrating distinct benefits such as stabilizing mitochondrial membrane potential (220).

Phenolic acids

Phenolic acids feature an aromatic ring substituted with ≥1 hydroxyl groups and a carboxyl group. The phenolic hydroxyls are primary sites for antioxidant activity via hydrogen atom transfer. The carboxyl group introduces an additional SAR dimension, allowing coordination with metal ions or formation of hydrogen bonds with target proteins (such as those involved in redox signaling), thereby stabilizing mitochondrial function and inhibiting NF-κB (221). In SIMD rats, phenolic acids efficiently scavenge ROS, preserve mitochondrial membrane potential, decrease intracellular Ca2+ overload and mitigate cardiomyocyte necrosis and apoptosis (128).

Alkaloids

Alkaloids contain ≥1 basic nitrogen atom within heterocyclic or polycyclic scaffolds. Examples relevant to SIMD include yohimbine and anisodamine. The basic nitrogen forms ionic interactions with acidic residues in receptors or enzymes, thereby modulating neurotransmitter release and inflammatory mediator levels (222). In sepsis, alkaloids often exhibit anti-inflammatory and anti-apoptotic effects by blocking α2-adrenergic receptors or inhibiting the NLRP3 inflammasome, helping to preserve cardiomyocyte function (199).

Saponins

Saponins are composed of triterpene or steroid aglycones linked to ≥1 sugar chains. This amphipathic structure defines their unique SAR: Aglycone enables interactions with and modulation of cell membrane properties and associated signaling pathways, while the sugar chains confer water solubility and influence pharmacokinetics (223). This structure allows saponins to modulate membrane fluidity and receptor clustering, leading to inhibition of caspase activation and decreased inflammatory mediator release, thereby mitigating apoptosis in cardiomyocytes (207).

Certain natural products display multi-target actions across SIMD-relevant pathways, combining anti-inflammatory and antioxidant effects with mitochondrial protection and anti-apoptotic signaling. Notable multi-target compounds include astragaloside IV, baicalein, carvacrol, curcumin, emodin, ginsenoside Re, matrine, propofol, quercetin and resveratrol (91,95,100,128,131). These agents exemplify the potential of pleiotropic natural products to regulate inflammation, oxidative stress, mitochondrial dysfunction and programmed cell death in SIMD (Table SI).

Notable therapeutic targets in SIMD

Key signaling pathways in SIMD

Natural products exert protective effects in SIMD by modulating multiple disease-associated pathways and molecular targets. Their primary actions are divided into four categories: Anti-inflammatory, antioxidant, anti-cell death and mitochondrial protection and repair (Fig. 7A). Notable signaling cascades include NF-κB, the MAPK family, the TLR4/MyD88 pathway, PI3K/Akt/mTOR, Nrf2/HO-1 and the NLRP3 inflammasome.

Notable therapeutic targets and
network pharmacology analysis in SIMD. (A) Primary actions of
natural products are categorized into anti-inflammatory,
antioxidant, anti-cell death and mitochondrial protection. Major
targeted signaling cascades include NF-κB, MAPK family, TLR4/MyD88,
PI3K/Akt/mTOR, Nrf2/HO-1 and the NLRP3 inflammasome. (B) Network
pharmacology comparison highlights high-frequency SIMD-associated
targets, such as iNOS (NOS2), NF-κB, MAPK members (JNK, ERK1/2),
PI3K/Akt/mTOR, NLRP3, SIRT1 and STAT3. SIMD, sepsis-induced
myocardial dysfunction; TLR, Toll like receptor; HO, heme
oxygenase; iNOS, inducible nitric oxide synthase ; SIRT, sirtuin
1.

Figure 7

Notable therapeutic targets and network pharmacology analysis in SIMD. (A) Primary actions of natural products are categorized into anti-inflammatory, antioxidant, anti-cell death and mitochondrial protection. Major targeted signaling cascades include NF-κB, MAPK family, TLR4/MyD88, PI3K/Akt/mTOR, Nrf2/HO-1 and the NLRP3 inflammasome. (B) Network pharmacology comparison highlights high-frequency SIMD-associated targets, such as iNOS (NOS2), NF-κB, MAPK members (JNK, ERK1/2), PI3K/Akt/mTOR, NLRP3, SIRT1 and STAT3. SIMD, sepsis-induced myocardial dysfunction; TLR, Toll like receptor; HO, heme oxygenase; iNOS, inducible nitric oxide synthase ; SIRT, sirtuin 1.

NF-κB serves as a key regulator of inflammation in SIMD (224). In cardiomyocytes and cardiac macrophages, LPS or endotoxin binding to TLR4/MyD88 activates the IKK complex, which phosphorylates and degrades IκBα. This frees NF-κB (p65/p50) to enter the nucleus and upregulate levels of IL-1β, IL-6, TNF-α and iNOS, thereby amplifying local inflammation and disturbing calcium homeostasis (225). As an upstream switch in early SIMD, TLR4 recruits TIR domain containing adaptor protein/MyD88 adaptors following LPS engagement, propagating both NF-κB and MAPK signaling and exacerbating myocardial injury (226,227).

The MAPK family, including p38, JNK, and ERK, also governs myocardial stress responses, inflammation and death in SIMD (228). p38 and JNK generally promote inflammation and apoptosis, while ERK supports survival or death depending on context (228). Overactivation of p38/JNK triggers c-Jun and ATF-2, leading to caspase-3-mediated apoptosis in SIMD (229). Nrf2/HO-1 provides the chief antioxidant defense. Under oxidative stress, Nrf2 dissociates from Keap1, translocates to the nucleus and binds antioxidant response elements to induce HO-1, NQO1 and other protective genes (230). Excess ROS in SIMD further damages membranes and amplifies NF-κB-driven inflammation (231).

PI3K/Akt/mTOR and SIRT1/AMPK form core networks that support cardiomyocyte survival and energy balance (232). PI3K activation leads to Akt phosphorylation and mTOR signaling, which promotes protein synthesis and restrains autophagy (233). SIRT1 deacetylates PGC-1α, p53 and NF-κB, coordinating mitochondrial biogenesis with anti-inflammatory effects (234). AMPK senses low ATP levels and works in concert with SIRT1 to maintain energy homeostasis (235). In SIMD, these pathways are typically downregulated, contributing to mitochondrial dysfunction and cell death (124).

Finally, the NLRP3 inflammasome and ferroptosis pathways drive inflammatory cell death in SIMD. NLRP3 assembly activates caspase-1, leading to IL-1β maturation and pyroptosis (236). Ferroptosis depends on iron-catalyzed lipid peroxidation, with key regulators including GPX4, acyl-CoA synthetase long chain family member 4 (ACSL4) and nuclear receptor coactivator 4) (237).

Rather than acting on a single target, natural products coordinate the aforementioned hubs to deliver combined anti-inflammatory, antioxidant, anti-death and mitochondrial-protective effects. Future studies should map the spatiotemporal dynamics and crosstalk between these pathways to guide the development of multitarget natural derivatives with improved precision and synergy.

Network pharmacology comparison of SIMD-associated targets

To evaluate how natural product targets align with known SIMD genes, 'sepsis', 'myocardial injury', and 'myocardial dysfunction' were in the Online Mendelian inheritance in Man and GeneCards databases (8,10). This search yielded over 350 unique disease-associated genes, with 41 common targets (Fig. 7B). High-frequency targets included iNOS (NOS2), NF-κB, MAPK family members (JNK, ERK1/2), PI3K/Akt/mTOR, NLRP3, SIRT1 and STAT3.

Notably, certain predicted targets, such as VEGF, cAMP, HSPs, Von Willebrand factor), MMP9 and PKC (Protein kinase C), lack thorough experimental validation in SIMD. Conversely, high-frequency targets such as NOX2, HO-1, GSK-3β, COX-2 and Nrf2 were not annotated as core SIMD targets in database queries. This discrepancy may reflect annotation lags, limited natural product-target interaction data and the inherent constraints of network pharmacology analyses. Integrating network pharmacology with systematic literature mining, multi-omics datasets and experimental validation is essential to build a comprehensive disease-drug-target network and inform the rational development of new SIMD therapies.

Docking of natural products to novel SIMD targets

To identify additional molecular targets for natural products in SIMD, molecular docking was performed for three proteins not yet validated in vitro or in vivo: cAMP-dependent protein kinase (cAMP), MMP9 and PKC). A total of five representative ligands (baicalein, quercetin, curcumin, matrine and resveratrol) were drawn in ChemDraw (14.0, revvitysignals. com/products/research/chemdraw), energy-minimized using the Molecular mechanics 2 force field, and docked to each target with AutoDock Vina (1.1.2, github.com/ccsb-scripps/AutoDock-Vina). The lowest-energy poses exhibiting the most favorable orientations were selected for analysis (238). All five compounds bound tightly to both MMP9 and PKC, with calculated binding energies <-8.0 kcal/mol (Fig. 8). Baicalein and curcumin also demonstrated notable affinity for PKA, with binding energies <-5.0 kcal/mol, suggesting potential direct modulation of this kinase. Key interacting residues and hydrogen-bond contacts are detailed in Table SII. These findings indicate that multitarget natural products may exert cardioprotective effects not only through established anti-inflammatory and antioxidant pathways but also by directly engaging cAMP-dependent protein kinase, MMP9 and PKC. This in silico evidence lays the groundwork for experimental validation of these novel interactions.

Docking of natural products to novel
sepsis-induced myocardial dysfunction) targets. All five compounds
bound tightly to both MMP9 and protein kinase C) (<-8.0
kcal/mol). Baicalein and curcumin also demonstrated appreciable
affinity for cAMP (<-5.0 kcal/mol).

Figure 8

Docking of natural products to novel sepsis-induced myocardial dysfunction) targets. All five compounds bound tightly to both MMP9 and protein kinase C) (<-8.0 kcal/mol). Baicalein and curcumin also demonstrated appreciable affinity for cAMP (<-5.0 kcal/mol).

Novel formulations for natural product therapeutics in SIMD

Although natural products offer multitarget synergy against SIMD, their clinical translation is hampered by poor solubility, low bioavailability and rapid clearance (239). Recent advances in formulation science, including nanotechnology, supramolecular chemistry and metal-organic coordination, provide new routes to improve delivery, targeting and efficacy (240).

Metal-organic nanozymes

Self-assembled nanozymes that incorporate metal-organic coordination combine drug delivery with enzyme-like catalysis (239,240). For example, ceria (CeO2) or iron oxide (Fe3O4) nanoparticles mimic superoxide dismutase and catalase through reversible Ce3+/Ce4+ or Fe2+/Fe3+ redox cycling, efficiently scavenging O2•- and H2O2 (241,242). By coordinating curcumin onto ceria, researchers have created ceria-curcumin hybrids (CeCHs) that exhibit dual SOD- and CAT-like activity in vitro (242). CeCHs neutralizes ROS, prevent glutathione peroxidase 4-induced ferroptosis in cardiomyocytes and shift macrophages toward an M2 phenotype (243). In LPS- and CLP-induced septic mice, CeCH decreases myocarditis and restores cardiac function (243). Similarly, Brazilin-Ce (IV) metal-organic nanoparticles inhibit IKKβ phosphorylation, suppress NF-κB signaling and deliver strong anti-inflammatory and cardioprotective effects in mice with myocardial infarction and sepsis (244).

Polymeric and lipid-based nanocarriers

Polymeric and lipid nanoparticles enhance the stability, circulation time and tissue distribution of natural products. Common polymers include poly(lactic-co-glycolic acid), PVA (Polyvinyl alcohol) and chitosan. Liposomal systems, solid lipid nanoparticles (SLNs) and mesoporous silica nanoparticles also serve as versatile carriers (245,246). In LPS-induced septic mice, curcumin-loaded SLNs decrease IL-6, TNF-α and IL-1β more effectively than free curcumin, while boosting IL-10 levels. This improvement is associated with stronger suppression of the TLR2/TLR4/NF-κB pathway and decreased multi-organ injury (247). Nano-curcumin further enhances mTOR pathway regulation and offers superior protection against myocardial ultrastructural damage in SIMD mice compared with the unformulated compound (248).

Cyclodextrin (CD)-based inclusion complexes

CD hosts form non-covalent inclusion complexes with hydrophobic drugs, improving their solubility and stability (249,250). Hydroxypropyl-β-CD (HPβCD) and methyl-β-CD (MβCD) are used (249). In neonatal mice with LPS-induced sepsis, naringenin/HPβCD complexes more effective than naringin in decreasing inflammatory cell infiltration in the lung, heart, kidney and brain. They also lower TNF-α, IL-1β and IL-6, increase IL-10, catalase and SOD activity and decrease lipid peroxidation and protein carbonylation, leading to improved survival rate (251). Quercetin/β-CD complexes extend plasma half-life of quercetin and enhance myocardial accumulation in CLP-induced septic rats, strengthening TLR4/NF-κB inhibition and improving cardiac function (252). Chemical derivatization, such as converting steviol glycosides into isosteviol sodium salt, further boosts water solubility and, in LPS-induced septic mice, enhances survival rate, multi-organ function and reduces inflammation and macrophage infiltration (253).

However, systematic pharmacokinetics and pharmacodynamics analyses, long-term toxicity studies and validation in large-animal models remain outstanding needs. Standardization of manufacturing methods, drug-loading efficiency and in vivo kinetics is also required. Future efforts should integrate multi-omics, high-resolution imaging and large-animal models to optimize formulations, clarify dosing regimens, define safety margins and accelerate clinical translation of these advanced delivery systems.

Discussion

Sepsis ranks as the third leading cause of death worldwide, affecting nearly 20 million people each year. A total of 40-60% of these patients develop SIMD, and their 28-day mortality is ~3 times higher than that of patients without cardiac involvement (11). This underscores the urgent need for effective, low-toxicity treatments. Natural products modulate key signaling networks, including TLR4/MyD88/NF-κB, MAPKs (p38/JNK/ERK), the NLRP3 inflammasome, PI3K/Akt/mTOR, SIRT1/AMPK and Nrf2/HO-1, to restore mitochondrial membrane potential, maintain ATP production and decrease inflammation, oxidative stress and cell death (207,217,220-222).

Multi-center randomized controlled trials indicate that formulations such as Xuebijing injection can decrease 28-day mortality and lower cardiac injury biomarkers (cTnI, NT-proBNP) and inflammatory markers (PCT), indicating systemic and possible myocardial protective effects (254,255). Similarly, the JinHong Formula decreases mortality and improves organ function scores, potentially through inhibition of inflammatory pathways such as IL-17 and TNF (256). Other formulations, including Dachaihu Tang and Shenfu injection, improve organ dysfunction scores, microcirculation and inflammatory or coagulation parameters (257,258). The alkaloid anisodamine has decreases serum lactate and improves mortality in septic shock (259,260). However, challenges remain. Most trials did not specifically enroll patients with confirmed SIMD, limiting direct conclusions about cardiac-specific efficacy. Furthermore, notable heterogeneity exists in study designs, sample size and the compositions of natural product formulations. Additionally, pharmacokinetic challenges constrain the use of natural products. Poor water solubility, low oral bioavailability and rapid systemic clearance all decrease therapeutic impact. To overcome these issues, researchers are developing novel formulations, such as polymeric or lipid nanoparticles, metal-organic frameworks, CD inclusion complexes, supramolecular assemblies and chemical derivatives, to improve solubility, stability, half-life and tissue targeting (261). Well-designed clinical trials are essential to assess safety, pharmacokinetics, pharmacodynamics and real-world efficacy in patients with SIMD.

Furthermore, a deeper understanding of the pathogenic network of SIMD is also needed. Extracellular vesicles and their miRNA cargo mediate crosstalk between immune cells and cardiomyocytes, regulating inflammation and apoptosis (262). Specific miRNAs fine-tune gene networks that control survival, fibrosis and autophagy (263). Endothelial-myocardial interactions, governed by adhesion molecules, inflammatory mediators and the glycocalyx, critically influence microcirculatory perfusion and cardiac stress responses (63,264). Mitochondria-endoplasmic reticulum contacts sites serve as hubs for calcium homeostasis, lipid metabolism and cell fate decisions (265,266). Additionally, emerging evidence highlights the role of the gut-heart axis in SIMD (267). Sepsis-induced disruption of the intestinal barrier leads to microbial translocation, systemic dissemination of PAMPs and elevated levels of gut-derived metabolites (267). This fuels a persistent systemic inflammatory state and may contribute directly to remote organ injury, including myocardial dysfunction (267). Conversely, beneficial microbial metabolites, such as short-chain fatty acids, modulate host immune responses and exert anti-inflammatory effects. Interventions targeting the gut microbiota or its metabolites hold promise for attenuating systemic inflammation and improving cardiac outcomes in sepsis, presenting a novel therapeutic avenue for SIMD (268). Thus, future efforts should leverage single-cell sequencing, organoid models and multi-omics approaches to build a comprehensive molecular map of SIMD and natural product interventions. Such integrative studies may identify new precision targets and accelerate the development of optimized, multitarget natural therapies for clinical use.

In conclusion, the present study provides a systematic overview of the structure-activity-mechanism relationships between natural products and SIMD. The mechanisms involve inhibition of the TLR4/MyD88-NF-κB/MAPK pathway and the NLRP3 inflammasome and activation of antioxidant and anti-apoptotic pathways, including PI3K/Akt/mTOR, SIRT1/AMPK and Nrf2/HO-1. The present review also maps the association between natural product classes and pharmacological activities and validated potential compounds against new SIMD targets via network pharmacology and molecular docking. Collectively, the present study clarified how natural product structures are associated with SIMD pathology and outlined a clear path from molecular design to clinical translation. The present study provides a systematic theoretical basis and practical avenues for multitarget natural product interventions in SIMD, with implications for drug discovery and clinical strategy advancement.

Supplementary Data

Availability of data and materials

The data generated in the present study are included in the figures and/or tables of this article.

Authors' contributions

FT contributed to study design, performed the literature review and drafted the manuscript. DL, SCZ and HMZ performed the literature review. XWQ supervised the study. All authors contributed to manuscript revision. All authors have read and approved the final manuscript. FT and XWQ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

HO-1

heme oxygenase-1

PAMP

pathogen-associated molecular pattern

PI3K

phosphoinositide 3-kinase

PKC

protein kinase C

SIMD

sepsis-induced myocardial dysfunction

SIRT1

sirtuin 1

Acknowledgements

Not applicable.

Funding

The present study was supported by Natural Science Foundation of Chongqing (grant no. CSTB2024NSCQ-KJFZZDX0023) and Chongqing Medical Scientific Research Project (Joint Project of Chongqing Health Commission and Science and Technology Bureau; grant no. 2022MSXM114).

References

1 

Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, et al: The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. 315:801–810. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Angus DC and van der Poll T: Severe sepsis and septic shock. N Engl J Med. 369:840–851. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Yan J, Li Z, Li Y and Zhang Y: Sepsis induced cardiotoxicity by promoting cardiomyocyte cuproptosis. Biochem Biophys Res Commun. 690:1492452024. View Article : Google Scholar

4 

Cecconi M, Evans L, Levy M and Rhodes A: Sepsis and septic shock. Lancet. 392:75–87. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Iyer S, Kennedy JN, Jentzer JC, Senussi MH and Seymour CW: Cardiac function before sepsis and clinical outcomes. JAMA. 331:1496–1499. 2024. View Article : Google Scholar : PubMed/NCBI

6 

van der Poll T and van Deventer SJ: Cytokines and anticytokines in the pathogenesis of sepsis. Infect Dis Clin North Am. 13:413–426. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Liu J, Li J, Tian P, Guli B, Weng G, Li L and Cheng Q: H2S attenuates sepsis-induced cardiac dysfunction via a PI3K/Akt-dependent mechanism. Exp Ther Med. 17:4064–4072. 2019.PubMed/NCBI

8 

Tang F, Zhang JN, Xu LY, Zhao XL, Wan F, Ao H and Peng C: Endothelial-derived exosomes: A novel therapeutic strategy for LPS-induced myocardial damage with anisodamine. Int J Biol Macromol. 282:1369932024. View Article : Google Scholar : PubMed/NCBI

9 

Tang X, Zhang C, Tian T, Dai X, Xing Y, Wang Y, Yang D, Li H, Wang Y, Lv X and Wang H: Posttreatment with dexmedetomidine aggravates LPS-induced myocardial dysfunction partly via activating cardiac endothelial α2A-AR in mice. Int Immunopharmacol. 116:1097242023. View Article : Google Scholar

10 

Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H and Peng C: Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res. 197:1069532023. View Article : Google Scholar : PubMed/NCBI

11 

Chen Y, Weng D, Shi W, Wei S, Ji W, Wang X, Xu Y, Wang X, Mei X and Guo S: Integrative network pharmacology and multi-omics reveal anisodamine hydrobromide's multi-target mechanisms in sepsis. Sci Rep. 15:279962025. View Article : Google Scholar : PubMed/NCBI

12 

Palmieri V, Innocenti F, Guzzo A, Guerrini E, Vignaroli D and Pini R: Left ventricular systolic longitudinal function as predictor of outcome in patients with sepsis. Circ Cardiovasc Imaging. 8:e0038652015. View Article : Google Scholar : PubMed/NCBI

13 

Fan R, Liu H and Liang Q: Roles and therapeutic targeting of exosomes in Sepsis-induced cardiomyopathy. J Cell Mol Med. 29:e705592025. View Article : Google Scholar : PubMed/NCBI

14 

Antonucci E, Fiaccadori E, Donadello K, Taccone FS, Franchi F and Scolletta S: Myocardial depression in sepsis: From pathogenesis to clinical manifestations and treatment. J Crit Care. 29:500–511. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Repessé X, Charron C and Vieillard-Baron A: Evaluation of left ventricular systolic function revisited in septic shock. Crit Care. 17:1642013. View Article : Google Scholar : PubMed/NCBI

16 

Aissaoui N, Boissier F, Chew M, Singer M and Vignon P: Sepsis-induced cardiomyopathy. Eur Heart J. 46:3339–3353. 2025. View Article : Google Scholar : PubMed/NCBI

17 

Parker MM, Shelhamer JH, Bacharach SL, Green MV, Natanson C, Frederick TM, Damske BA and Parrillo JE: Profound but reversible myocardial depression in patients with septic shock. Ann Intern Med. 100:483–490. 1984. View Article : Google Scholar : PubMed/NCBI

18 

Zaky A, Deem S, Bendjelid K and Treggiari MM: Characterization of cardiac dysfunction in sepsis: An ongoing challenge. Shock. 41:12–24. 2014. View Article : Google Scholar

19 

ver Elst KM, Spapen HD, Nguyen DN, Garbar C, Huyghens LP and Gorus FK: Cardiac troponins I and T are biological markers of left ventricular dysfunction in septic shock. Clin Chem. 46:650–657. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Sheyin O, Davies O, Duan W and Perez X: The prognostic significance of troponin elevation in patients with sepsis: A meta-analysis. Heart Lung. 44:75–81. 2015. View Article : Google Scholar

21 

Clerico A, Iervasi G and Mariani G: Pathophysiologic relevance of measuring the plasma levels of cardiac natriuretic peptide hormones in humans. Horm Metab Res. 31:487–498. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Chua G and Kang-Hoe L: Marked elevations in N-terminal brain natriuretic peptide levels in septic shock. Crit Care. 8:R248–R250. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Roch A, Allardet-Servent J, Michelet P, Oddoze C, Forel JM, Barrau K, Loundou A, Perrin G, Auffray JP, Portugal H and Papazian L: NH2 terminal pro-brain natriuretic peptide plasma level as an early marker of prognosis and cardiac dysfunction in septic shock patients. Crit Care Med. 33:1001–1007. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Alam ML, Katz R, Bellovich KA, Bhat ZY, Brosius FC, de Boer IH, Gadegbeku CA, Gipson DS, Hawkins JJ, Himmelfarb J, et al: Soluble ST2 and Galectin-3 and Progression of CKD. Kidney Int Rep. 4:103–111. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Chang X, Guo Y, Wang J, Liu J, Ma Y, Lu Q and Han Y: Heart-type fatty acid binding protein (H-FABP) as an early biomarker in sepsis-induced cardiomyopathy: A prospective observational study. Lipids Health Dis. 23:2832024. View Article : Google Scholar : PubMed/NCBI

26 

Weinberger J, Klompas M and Rhee C: What is the utility of measuring lactate levels in patients with sepsis and septic shock? Semin Respir Crit Care Med. 42:650–661. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Benz F, Roy S, Trautwein C, Roderburg C and Luedde T: Circulating MicroRNAs as biomarkers for sepsis. Int J Mol Sci. 17:782016. View Article : Google Scholar : PubMed/NCBI

28 

Manetti AC, Maiese A, Paolo MD, De Matteis A, La Russa R, Turillazzi E, Frati P and Fineschi V: MicroRNAs and Sepsis-induced cardiac dysfunction: A systematic review. Int J Mol Sci. 22:3212020. View Article : Google Scholar

29 

Ketelut-Carneiro N and Fitzgerald KA: Apoptosis, pyroptosis, and Necroptosis-Oh My! The many ways a cell can die. J Mol Biol. 434:1673782022. View Article : Google Scholar

30 

Communal C, Sumandea M, de Tombe P, Narula J, Solaro RJ and Hajjar RJ: Functional consequences of caspase activation in cardiac myocytes. Proc Natl Acad Sci USA. 99:6252–6256. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Nevière R, Fauvel H, Chopin C, Formstecher P and Marchetti P: Caspase inhibition prevents cardiac dysfunction and heart apoptosis in a rat model of sepsis. Am J Respir Crit Care Med. 163:218–225. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Hu H, Tian M, Ding C and Yu S: The C/EBP Homologous protein (CHOP) Transcription factor functions in endoplasmic reticulum Stress-induced apoptosis and microbial infection. Front Immunol. 9:30832018. View Article : Google Scholar

33 

Li L, Peng X, Guo L, Zhao Y and Cheng Q: Sepsis causes heart injury through endoplasmic reticulum stress-mediated apoptosis signaling pathway. Int J Clin Exp Pathol. 13:964–971. 2020.PubMed/NCBI

34 

Xu X, Liu Q, He S, Zhao J, Wang N, Han X and Guo Y: Qiang-Xin 1 formula prevents Sepsis-induced apoptosis in murine cardiomyocytes by suppressing endoplasmic Reticulum- and Mitochondria-associated pathways. Front Pharmacol. 9:8182018. View Article : Google Scholar : PubMed/NCBI

35 

Zheng X, Chen W, Gong F, Chen Y and Chen E: The role and mechanism of pyroptosis and potential therapeutic targets in sepsis: A review. Front Immunol. 12:7119392021. View Article : Google Scholar : PubMed/NCBI

36 

Xue Z, Xi Q, Liu H, Guo X, Zhang J, Zhang Z, Li Y, Yang G, Zhou D, Yang H, et al: miR-21 promotes NLRP3 inflammasome activation to mediate pyroptosis and endotoxic shock. Cell Death Dis. 10:4612019. View Article : Google Scholar : PubMed/NCBI

37 

Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, et al: Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 116:2672–2680. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Li W, Li W, Leng Y, Xiong Y and Xia Z: Ferroptosis is involved in diabetes myocardial Ischemia/reperfusion injury through endoplasmic reticulum stress. DNA Cell Biol. 39:210–225. 2020. View Article : Google Scholar

39 

Parzych KR and Klionsky DJ: An overview of autophagy: Morphology, mechanism, and regulation. Antioxid Redox Signal. 20:460–473. 2014. View Article : Google Scholar :

40 

Liu AB, Li SJ, Yu YY, Zhang JF and Ma L: Current insight on the mechanisms of programmed cell death in sepsis-induced myocardial dysfunction. Front Cell Dev Biol. 11:13097192023. View Article : Google Scholar :

41 

Denk S, Perl M and Huber-Lang M: Damage- and pathogen-associated molecular patterns and alarmins: Keys to sepsis? Eur Surg Res. 48:171–179. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Vénéreau E, Ceriotti C and Bianchi ME: DAMPs from cell death to new life. Front Immunol. 6:4222015. View Article : Google Scholar : PubMed/NCBI

43 

Hobai IA, Morse JC, Siwik DA and Colucci WS: Lipopolysaccharide and cytokines inhibit rat cardiomyocyte contractility in vitro. J Surg Res. 193:888–901. 2015. View Article : Google Scholar

44 

Zhang YY and Ning BT: Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther. 6:4072021. View Article : Google Scholar : PubMed/NCBI

45 

Fujimura K, Karasawa T, Komada T, Yamada N, Mizushina Y, Baatarjav C, Matsumura T, Otsu K, Takeda N, Mizukami H, et al: NLRP3 inflammasome-driven IL-1β and IL-18 contribute to lipopolysaccharide-induced septic cardiomyopathy. J Mol Cell Cardiol. 180:58–68. 2023. View Article : Google Scholar : PubMed/NCBI

46 

Busch K, Kny M, Huang N, Klassert TE, Stock M, Hahn A, Graeger S, Todiras M, Schmidt S, Chamling B, et al: Inhibition of the NLRP3/IL-1β axis protects against sepsis-induced cardiomyopathy. J Cachexia Sarcopenia Muscle. 12:1653–1668. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Kumar V: Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int Immunopharmacol. 89:1070872020. View Article : Google Scholar : PubMed/NCBI

48 

Hoover DB, Ozment TR, Wondergem R, Li C and Williams DL: Impaired heart rate regulation and depression of cardiac chronotropic and dromotropic function in polymicrobial sepsis. Shock. 43:185–191. 2015. View Article : Google Scholar :

49 

Nolfi-Donegan D, Braganza A and Shiva S: Mitochondrial electron transport chain: Oxidative phosphorylation, oxidant production, and methods of measurement. Redox Biol. 37:1016742020. View Article : Google Scholar : PubMed/NCBI

50 

Chen YR and Zweier JL: Cardiac mitochondria and reactive oxygen species generation. Circ Res. 114:524–537. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Sun F, Huo X, Zhai Y, Wang A, Xu J, Su D, Bartlam M and Rao Z: Crystal structure of mitochondrial respiratory membrane protein complex II. Cell. 121:1043–1057. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Tsolaki V, Makris D, Mantzarlis K and Zakynthinos E: Sepsis-induced cardiomyopathy: Oxidative implications in the initiation and resolution of the damage. Oxid Med Cell Longev. 2017:73935252017. View Article : Google Scholar : PubMed/NCBI

53 

Chen YR, Chen CL, Yeh A, Liu X and Zweier JL: Direct and indirect roles of cytochrome b in the mediation of superoxide generation and NO catabolism by mitochondrial succinate-cytochrome c reductase. J Biol Chem. 281:13159–13168. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Takasu O, Gaut JP, Watanabe E, To K, Fagley RE, Sato B, Jarman S, Efimov IR, Janks DL, Srivastava A, et al: Mechanisms of cardiac and renal dysfunction in patients dying of sepsis. Am J Respir Crit Care Med. 187:509–517. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Vanasco V, Saez T, Magnani ND, Pereyra L, Marchini T, Corach A, Vaccaro MI, Corach D, Evelson P and Alvarez S: Cardiac mitochondrial biogenesis in endotoxemia is not accompanied by mitochondrial function recovery. Free Radic Biol Med. 77:1–9. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Haileselassie B, Mukherjee R, Joshi AU, Napier BA, Massis LM, Ostberg NP, Queliconi BB, Monack D, Bernstein D and Mochly-Rosen D: Drp1/Fis1 interaction mediates mitochondrial dysfunction in septic cardiomyopathy. J Mol Cell Cardiol. 130:160–169. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Wagner S, Schürmann S, Hein S, Schüttler J and 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. 110:5072015. View Article : Google Scholar : PubMed/NCBI

58 

Lin Y, Xu Y and Zhang Z: Sepsis-induced myocardial dysfunction (SIMD): The pathophysiological mechanisms and therapeutic strategies targeting mitochondria. Inflammation. 43:1184–1200. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Ravikumar N, Sayed MA, Poonsuph CJ, Sehgal R, Shirke MM and Harky A: Septic cardiomyopathy: From basics to management choices. Curr Probl Cardiol. 46:1007672021. View Article : Google Scholar : PubMed/NCBI

60 

Cao T, Ni R, Ding W, Ji X, Fan GC, Zhang Z and Peng T: Nicotinamide mononucleotide as a therapeutic agent to alleviate multi-organ failure in sepsis. J Transl Med. 21:8832023. View Article : Google Scholar : PubMed/NCBI

61 

Furian T, Aguiar C, Prado K, Ribeiro RV, Becker L, Martinelli N, Clausell N, Rohde LE and Biolo A: Ventricular dysfunction and dilation in severe sepsis and septic shock: Relation to endothelial function and mortality. J Crit Care. 27:319.e9–e15. 2012. View Article : Google Scholar

62 

Kacimi R, Karliner JS, Koudssi F and Long CS: Expression and regulation of adhesion molecules in cardiac cells by cytokines: Response to acute hypoxia. Circ Res. 82:576–586. 1998. View Article : Google Scholar : PubMed/NCBI

63 

Tang F, Zhao XL, Xu LY, Zhang JN, Ao H and Peng C: Endothelial dysfunction: Pathophysiology and therapeutic targets for sepsis-induced multiple organ dysfunction syndrome. Biomed Pharmacother. 178:1171802024. View Article : Google Scholar : PubMed/NCBI

64 

Zhan JH, Wei J, Liu YJ, Wang PX and Zhu XY: Sepsis-associated endothelial glycocalyx damage: A review of animal models, clinical evidence, and molecular mechanisms. Int J Biol Macromol. 295:1395482025. View Article : Google Scholar : PubMed/NCBI

65 

Burg N, Malpass R, Alex L, Tran M, Englebrecht E, Kuo A, Pannelini T, Minett M, Athukorala K and Worgall T: Endothelial cell sphingosine 1-phosphate receptor 1 restrains VE-cadherin cleavage and attenuates experimental inflammatory arthritis. JCI Insight. 9:e1714672024. View Article : Google Scholar : PubMed/NCBI

66 

de Oliveira J and Miranda CH: Doxycycline protects against sepsis-induced endothelial glycocalyx shedding. Sci Rep. 14:104772024. View Article : Google Scholar : PubMed/NCBI

67 

van de Sandt AM, Windler R, Gödecke A, Ohlig J, Zander S, Reinartz M, Graf J, van Faassen EE, Rassaf T, Schrader J, et al: Endothelial NOS (NOS3) impairs myocardial function in developing sepsis. Basic Res Cardiol. 108:3302013. View Article : Google Scholar : PubMed/NCBI

68 

Zeng N, Xu J, Yao W, Li S, Ruan W and Xiao F: Brain-derived neurotrophic factor attenuates septic myocardial dysfunction via eNOS/NO pathway in rats. Oxid Med Cell Longev. 2017:17214342017. View Article : Google Scholar : PubMed/NCBI

69 

Hong G, Zheng D, Zhang L, Ni R, Wang G, Fan GC, Lu Z and Peng T: Administration of nicotinamide riboside prevents oxidative stress and organ injury in sepsis. Free Radic Biol Med. 123:125–137. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Mirna M, Paar V, Rezar R, Topf A, Eber M, Hoppe UC, Lichtenauer M and Jung C: MicroRNAs in inflammatory heart diseases and Sepsis-induced cardiac dysfunction: A potential scope for the future? Cells. 8:13522019. View Article : Google Scholar : PubMed/NCBI

71 

Gao M, Wang X, Zhang X, Ha T, Ma H, Liu L, Kalbfleisch JH, Gao X, Kao RL, Williams DL and Li C: Attenuation of cardiac dysfunction in polymicrobial sepsis by MicroRNA-146a is mediated via targeting of IRAK1 and TRAF6 expression. J Immunol. 195:672–682. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Ma H, Wang X, Ha T, Gao M, Liu L, Wang R, Yu K, Kalbfleisch JH, Kao RL, Williams DL and Li C: MicroRNA-125b prevents cardiac dysfunction in polymicrobial sepsis by targeting TRAF6-Mediated nuclear factor κB activation and p53-Mediated apoptotic signaling. J Infect Dis. 214:1773–1783. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Yao Y, Sun F and Lei M: miR-25 inhibits sepsis-induced cardiomyocyte apoptosis by targetting PTEN. Biosci Rep. 38:BSR201715112018. View Article : Google Scholar : PubMed/NCBI

74 

Li Z, Yi N, Chen R, Meng Y, Wang Y, Liu H, Cao W, Hu Y, Gu Y, Tong C, et al: miR-29b-3p protects cardiomyocytes against endotoxin-induced apoptosis and inflammatory response through targeting FOXO3A. Cell Signal. 74:1097162020. View Article : Google Scholar : PubMed/NCBI

75 

Long X, Huang Y, He J, Zhang X, Zhou Y, Wei Y, Tang Y and Liu L: Upregulation of miR-335 exerts protective effects against sepsis-induced myocardial injury. Mol Med Rep. 24:8062021. View Article : Google Scholar :

76 

Liang L, Liu S, Wu Q, Chen R, Jiang S and Yang Z: m6A-mediated upregulation of miRNA-193a aggravates cardiomyocyte apoptosis and inflammatory response in sepsis-induced cardiomyopathy via the METTL3/miRNA-193a/BCL2L2 pathway. Exp Cell Res. 430:1137122023. View Article : Google Scholar

77 

He Z, Xu L, Zeng X, Yang B, Liu P, Han D, Xue H and Luo B: circROCK1 Promotes septic myocardial injury through regulating miR-96-5p/OXSR1 axis. Acta Biochim Pol. 70:567–574. 2023.PubMed/NCBI

78 

Wang H, Bei Y, Shen S, Huang P, Shi J, Zhang J, Sun Q, Chen Y, Yang Y, Xu T, et al: miR-21-3p controls sepsis-associated cardiac dysfunction via regulating SORBS2. J Mol Cell Cardiol. 94:43–53. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Ge C, Liu J and Dong S: miRNA-214 protects Sepsis-induced myocardial injury. Shock. 50:112–118. 2018. View Article : Google Scholar

80 

Li Y, Sun G and Wang L: MiR-21 participates in LPS-induced myocardial injury by targeting Bcl-2 and CDK6. Inflamm Res. 71:205–214. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Zhu XG, Zhang TN, Wen R and Liu CF: Overexpression of miR-150-5p alleviates apoptosis in Sepsis-induced myocardial depression. Biomed Res Int. 2020:30231862020. View Article : Google Scholar : PubMed/NCBI

82 

Zhang L, Li B, Li W, Jiang J, Chen W, Yang H and Pan D: miR-107 attenuates Sepsis-induced myocardial injury by targeting PTEN and activating the PI3K/AKT signaling pathway. Cells Tissues Organs. 212:523–534. 2023. View Article : Google Scholar

83 

Sun F, Yuan W, Wu H, Chen G, Sun Y, Yuan L, Zhang W and Lei M: LncRNA KCNQ1OT1 attenuates sepsis-induced myocardial injury via regulating miR-192-5p/XIAP axis. Exp Biol Med (Maywood). 245:620–630. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Chen DD, Wang HW and Cai XJ: Long non-coding RNA ZFAS1 alleviates sepsis-induced myocardial injury via target miR-34b-5p/SIRT1. Innate Immun. 27:377–387. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Xu LJ, Yang Y, Yuan LF, Liu H, Xu NP, Yang Y and Huang L: SP1-stimulated miR-208a-5p aggravates sepsis-induced myocardial injury via targeting XIAP. Exp Cell Res. 435:1139052024. View Article : Google Scholar : PubMed/NCBI

86 

Li Y, Shao Y, Su J and Dong S: MiR-383-3p attenuates sepsis-induced myocardial ferroptosis by targeting ATF4 and inhibiting the ATF4-CHOP-CHAC1 signaling axis. Cell Signal. 136:1121692025. View Article : Google Scholar : PubMed/NCBI

87 

Wang J, Wei T, Zhang W, Chu Y, Zhang D, Zhang M, Hu J, Ji Z and Hao Q: Inhibition of miR-194-5p avoids DUSP9 downregulation thus limiting sepsis-induced cardiomyopathy. Sci Rep. 14:203132024. View Article : Google Scholar : PubMed/NCBI

88 

Liang D, Jin Y, Lin M, Xia X, Chen X and Huang A: Down-regulation of Xist and Mir-7a-5p improves LPS-induced myocardial injury. Int J Med Sci. 17:2570–2577. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Gong M, Tao L and Li X: MicroRNA-21-3p/Rcan1 signaling axis affects apoptosis of cardiomyocytes of sepsis rats. Gen Physiol Biophys. 42:217–227. 2023. View Article : Google Scholar : PubMed/NCBI

90 

Dao L, Liu H, Xiu R, Yao T, Tong R and Xu L: Gramine improves sepsis-induced myocardial dysfunction by binding to NF-κB p105 and inhibiting its ubiquitination. Phytomedicine. 125:1553252024. View Article : Google Scholar

91 

Chen D, Wang H and Cai X: Curcumin interferes with sepsis-induced cardiomyocyte apoptosis via TLR1 inhibition. Rev Port Cardiol. 42:209–221. 2023.In English, Portuguese. View Article : Google Scholar : PubMed/NCBI

92 

Zhang J, Zhu D, Wang Y and Ju Y: Andrographolide attenuates LPS-induced cardiac malfunctions through Inhibition of IκB phosphorylation and apoptosis in mice. Cell Physiol Biochem. 37:1619–1628. 2015. View Article : Google Scholar

93 

Wang YY, Li HM, Wang HD, Peng XM, Wang YP, Lu DX, Qi RB, Hu CF and Jiang JW: Pretreatment with berberine and yohimbine protects against LPS-induced myocardial dysfunction via inhibition of cardiac I-[kappa]B[alpha] phosphorylation and apoptosis in mice. Shock. 35:322–328. 2011. View Article : Google Scholar

94 

Meng YY, Liu Y, Hu ZF, Zhang Y, Ni J, Ma ZG, Liao HH, Wu QQ and Tang QZ: Sanguinarine attenuates lipopolysaccharide-induced inflammation and apoptosis by inhibiting the TLR4/NF-κB pathway in H9c2 Cardiomyocytes. Curr Med Sci. 38:204–211. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Zhong L, Zhou XL, Liu YS, Wang YM, Ma F, Guo BL, Yan ZQ and Zhang QY: Estrogen receptor α mediates the effects of notoginsenoside R1 on endotoxin-induced inflammatory and apoptotic responses in H9c2 cardiomyocytes. Mol Med Rep. 12:119–126. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Sun B, Xiao J, Sun XB and Wu Y: Notoginsenoside R1 attenuates cardiac dysfunction in endotoxemic mice: An insight into oestrogen receptor activation and PI3K/Akt signalling. Br J Pharmacol. 168:1758–1770. 2013. View Article : Google Scholar :

97 

Zhu H, Zhang L, Jia H, Xu L, Cao Y, Zhai M, Li K, Xia L, Jiang L, Li X, et al: Tetrahydrocurcumin improves lipopolysaccharide-induced myocardial dysfunction by inhibiting oxidative stress and inflammation via JNK/ERK signaling pathway regulation. Phytomedicine. 104:1542832022. View Article : Google Scholar : PubMed/NCBI

98 

Xie WJ, Hou G, Wang L, Wang SS and Xiong XX: Astaxanthin suppresses lipopolysaccharide-induced myocardial injury by regulating MAPK and PI3K/AKT/mTOR/GSK3β signaling. Mol Med Rep. 22:3338–3346. 2020.PubMed/NCBI

99 

Jiang L, Zhang L, Yang J, Shi H, Zhu H, Zhai M, Lu L, Wang X, Li XY, Yu S, et al: 1-Deoxynojirimycin attenuates septic cardiomyopathy by regulating oxidative stress, apoptosis, and inflammation via the JAK2/STAT6 signaling pathway. Biomed Pharmacother. 155:1136482022. View Article : Google Scholar : PubMed/NCBI

100 

Su Y, Yin X, Huang X, Guo Q, Ma M and Guo L: Astragaloside IV ameliorates sepsis-induced myocardial dysfunction by regulating NOX4/JNK/BAX pathway. Life Sci. 310:1211232022. View Article : Google Scholar : PubMed/NCBI

101 

Zhang T, Yan T, Du J, Wang S and Yang H: Apigenin attenuates heart injury in lipopolysaccharide-induced endotoxemic model by suppressing sphingosine kinase 1/sphingosine 1-phosphate signaling pathway. Chem Biol Interact. 233:46–55. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Yu H, Du Q, Wu J, Feng F, Hou S, Liu M, Wang S, Liu X, Wang C and Xu K: Gastrodin regulates H3K14la through the CDT2-KAT2A axis to treat sepsis-induced myocardial dysfunction. Int Immunopharmacol. 161:1150652025. View Article : Google Scholar : PubMed/NCBI

103 

Wang RY, Wang MG, Tang HZ, Du H, Luo Y, Li Q, Zhang XH, Fu J and Lv CZ: The protective effects of ruscogenin against Lipopolysaccharide-induced myocardial injury in septic mice. J Cardiovasc Pharmacol. 84:175–187. 2024. View Article : Google Scholar : PubMed/NCBI

104 

Zhu XX, Meng XY, Zhang AY, Zhao CY, Chang C, Chen TX, Huang YB, Xu JP, Fu X, Cai WW, et al: Vaccarin alleviates septic cardiomyopathy by potentiating NLRP3 palmitoylation and inactivation. Phytomedicine. 131:1557712024. View Article : Google Scholar : PubMed/NCBI

105 

Long H, Xu B, Luo Y and Luo K: Artemisinin protects mice against burn sepsis through inhibiting NLRP3 inflammasome activation. Am J Emerg Med. 34:772–777. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Liu H, Sun Y, Zhang Y, Yang G, Guo L, Zhao Y and Pei Z: Role of thymoquinone in cardiac damage caused by sepsis from BALB/c mice. Inflammation. 42:516–525. 2019. View Article : Google Scholar

107 

Wei A, Liu J, Li D, Lu Y, Yang L, Zhuo Y, Tian W and Cong H: Syringaresinol attenuates sepsis-induced cardiac dysfunction by inhibiting inflammation and pyroptosis in mice. Eur J Pharmacol. 913:1746442021. View Article : Google Scholar : PubMed/NCBI

108 

Luo M, Yan D, Sun Q, Tao J, Xu L, Sun H and Zhao H: Ginsenoside Rg1 attenuates cardiomyocyte apoptosis and inflammation via the TLR4/NF-κB/NLRP3 pathway. J Cell Biochem. 121:2994–3004. 2020. View Article : Google Scholar

109 

Shao F, Zhou L, Zhang Y, Chen H, Zhang Y and Guan Z: Gastrodin alleviates inflammatory injury of cardiomyocytes in septic shock mice via inhibiting NLRP3 expression. In Vitro Cell Dev Biol Anim. 57:571–581. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Khodir AE, Samra YA and Said E: A novel role of nifuroxazide in attenuation of sepsis-associated acute lung and myocardial injuries; role of TLR4/NLPR3/IL-1β signaling interruption. Life Sci. 256:1179072020. View Article : Google Scholar

111 

Song P, Shen DF, Meng YY, Kong CY, Zhang X, Yuan YP, Yan L, Tang QZ and Ma ZG: Geniposide protects against sepsis-induced myocardial dysfunction through AMPKα-dependent pathway. Free Radic Biol Med. 152:186–196. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Dai S, Ye B, Chen L, Hong G, Zhao G and Lu Z: Emodin alleviates LPS-induced myocardial injury through inhibition of NLRP3 inflammasome activation. Phytother Res. 35:5203–5213. 2021. View Article : Google Scholar : PubMed/NCBI

113 

Du R, Yun Q, Wang Y, Dou X, Ye H, Wang J and Gao Q: Plumbagin protect against sepsis-induced myocardial injury in mice by inhibiting the JAK2/STAT3 signaling pathway to reduce cardiomyocyte pyroptosis. Nan Fang Yi Ke Da Xue Xue Bao. 44:2209–2219. 2024.In Chinese. PubMed/NCBI

114 

Joshi S, Kundu S, Priya VV, Kulhari U, Mugale MN and Sahu BD: Anti-inflammatory activity of carvacrol protects the heart from lipopolysaccharide-induced cardiac dysfunction by inhibiting pyroptosis via NLRP3/Caspase1/Gasdermin D signaling axis. Life Sci. 324:1217432023. View Article : Google Scholar : PubMed/NCBI

115 

Wang Y, Feng W, Li S, Liu C, Jia L, Wang P, Li L, Du H and Yu W: Oxycodone attenuates lipopolysaccharide-induced myocardial injury by inhibiting inflammation, oxidation and pyroptosis via Nrf2/HO-1 signalling pathway. Clin Exp Pharmacol Physiol. 51:e139102024. View Article : Google Scholar : PubMed/NCBI

116 

Wu B, Song H, Fan M, You F, Zhang L, Luo J, Li J, Wang L, Li C and Yuan M: Luteolin attenuates sepsis-induced myocardial injury by enhancing autophagy in mice. Int J Mol Med. 45:1477–1487. 2020.PubMed/NCBI

117 

Shiroorkar PN, Afzal O, Kazmi I, Al-Abbasi FA, Altamimi ASA, Gubbiyappa KS and Sreeharsha N: Cardioprotective effect of tangeretin by inhibiting PTEN/AKT/mTOR axis in experimental Sepsis-induced myocardial dysfunction. Molecules. 25:56222020. View Article : Google Scholar : PubMed/NCBI

118 

Cardenas H, Arango D, Nicholas C, Duarte S, Nuovo GJ, He W, Voss OH, Gonzalez-Mejia ME, Guttridge DC, Grotewold E and Doseff AI: Dietary apigenin exerts Immune-regulatory activity in vivo by reducing NF-κB activity, halting leukocyte infiltration and restoring normal metabolic function. Int J Mol Sci. 17:3232016. View Article : Google Scholar

119 

Li F, Lang F, Zhang H, Xu L, Wang Y, Zhai C and Hao E: Apigenin alleviates Endotoxin-induced myocardial toxicity by modulating inflammation, oxidative stress, and autophagy. Oxid Med Cell Longev. 2017:23028962017. View Article : Google Scholar : PubMed/NCBI

120 

Chang X, He Y, Wang L, Luo C, Liu Y and Li R: Puerarin alleviates LPS-induced H9C2 cell injury by inducing mitochondrial autophagy. J Cardiovasc Pharmacol. 80:600–608. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Tang R, Jia L, Li Y, Zheng J and Qi P: Narciclasine attenuates sepsis-induced myocardial injury by modulating autophagy. Aging (Albany NY). 13:15151–15163. 2021. View Article : Google Scholar : PubMed/NCBI

122 

Yuan X, Chen G, Guo D, Xu L and Gu Y: Polydatin alleviates septic myocardial injury by promoting SIRT6-mediated autophagy. Inflammation. 43:785–795. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Yu YW, Chen X, Yan JY, Hu J, Huang KY, Ji KT and Cai HL: Phlorizin, a novel caloric restriction mimetic, stimulates hypoxia and protects cardiomyocytes through activating autophagy via modulating the Hif-1α/Bnip3 axis in sepsis-induced myocardial dysfunction. Int Immunopharmacol. 126:1112412024. View Article : Google Scholar

124 

Zhou B, Zhang J, Chen Y, Liu Y, Tang X, Xia P, Yu P and Yu S: Puerarin protects against sepsis-induced myocardial injury through AMPK-mediated ferroptosis signaling. Aging (Albany NY). 14:3617–3632. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Lin X, Zhao X, Chen Q, Wang X, Wu Y and Zhao H: Quercetin ameliorates ferroptosis of rat cardiomyocytes via activation of the SIRT1/p53/SLC7A11 signaling pathway to alleviate sepsis-induced cardiomyopathy. Int J Mol Med. 52:1162023. View Article : Google Scholar :

126 

Xiao Y, Yu Y, Hu L, Yang Y, Yuan Y, Zhang W, Luo J and Yu L: Matrine alleviates Sepsis-induced myocardial injury by inhibiting ferroptosis and apoptosis. Inflammation. 46:1684–1696. 2023. View Article : Google Scholar : PubMed/NCBI

127 

Lin LQ, Mao FK, Lin J, Guo L, Yuan WR and Wang BY: Ginsenoside Rg1 induces ferroptosis by regulating the focal adhesion kinase/protein kinase B-forkhead box O3A signaling pathway and alleviates sepsis-induced myocardial damage. J Physiol Pharmacol. 75:2024. View Article : Google Scholar

128 

Wang X, Simayi A, Fu J, Zhao X and Xu G: Resveratrol mediates the miR-149/HMGB1 axis and regulates the ferroptosis pathway to protect myocardium in endotoxemia mice. Am J Physiol Endocrinol Metab. 323:e21–e32. 2022. View Article : Google Scholar : PubMed/NCBI

129 

Tang R, Jiang M, Tang X, Chen S, Xu H, Pan Y, Lin B, Wei X, Ye Q, Wu M and Qi P: Narciclasine mitigates sepsis-induced cardiac dysfunction by enhancing BNIP3-mediated mitophagy and suppressing ferroptosis. Free Radic Biol Med. 238:220–234. 2025. View Article : Google Scholar : PubMed/NCBI

130 

Zeng Y, Cao G, Lin L, Zhang Y, Luo X, Ma X, Aiyisake A and Cheng Q: Resveratrol attenuates Sepsis-induced cardiomyopathy in rats through Anti-Ferroptosis via the Sirt1/Nrf2 pathway. J Invest Surg. 36:21575212023. View Article : Google Scholar

131 

Ye H, Wu L, Liu YM, Zhang JX, Hu HT, Dong ML and Ren J: Wogonin attenuates septic cardiomyopathy by suppressing ALOX15-mediated ferroptosis. Acta Pharmacol Sin. 46:2407–2422. 2025. View Article : Google Scholar : PubMed/NCBI

132 

Guan F, Du H, Li J, Ren H and Dong A: Quercetin alleviates LPS-stimulated myocardial injury through regulating ALOX5/PI3K/AKT pathway in sepsis. Cardiovasc Toxicol. 24:1116–1124. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Huang SH, Xu M, Wu HM, Wan CX, Wang HB, Wu QQ, Liao HH, Deng W and Tang QZ: Isoquercitrin attenuated cardiac dysfunction via AMPKα-Dependent pathways in LPS-Treated mice. Mol Nutr Food Res. 62:e18009552018. View Article : Google Scholar

134 

Wei X, Meng X, Yuan Y, Shen F, Li C and Yang J: Quercetin exerts cardiovascular protective effects in LPS-induced dysfunction in vivo by regulating inflammatory cytokine expression, NF-κB phosphorylation, and caspase activity. Mol Cell Biochem. 446:43–52. 2018. View Article : Google Scholar : PubMed/NCBI

135 

Zhou Q, Zeng X, Kang W, Pan X, Wang L and Xia Z: Ciprofol attenuates sepsis-induced cardiomyopathy via α7 nicotinic acetylcholine receptor-dependent modulation of myocardial inflammation and NF-κB/STAT3 signaling. Eur J Pharmacol. 1003:1779832025. View Article : Google Scholar

136 

Huang X, Zhang MZ, Liu B, Ma SY, Yin X and Guo LH: Astragaloside IV Attenuates polymicrobial Sepsis-induced cardiac dysfunction in rats via IKK/NF-κB pathway. Chin J Integr Med. 27:825–831. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Chen S and Fan B: Myricetin protects cardiomyocytes from LPS-induced injury. Herz. 43:265–274. 2018. View Article : Google Scholar

138 

Zhang N, Feng H, Liao HH, Chen S, Yang Z, Deng W and Tang QZ: Myricetin attenuated LPS induced cardiac injury in vivo and in vitro. Phytother Res. 32:459–470. 2018. View Article : Google Scholar

139 

Xianchu L, Lan PZ, Qiufang L, Yi L, Xiangcheng R, Wenqi H and Yang D: Naringin protects against lipopolysaccharide-induced cardiac injury in mice. Environ Toxicol Pharmacol. 48:1–6. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Sun LJ, Qiao W, Xiao YJ, Cui L, Wang X and Ren WD: Naringin mitigates myocardial strain and the inflammatory response in sepsis-induced myocardial dysfunction through regulation of PI3K/AKT/NF-κB pathway. Int Immunopharmacol. 75:1057822019. View Article : Google Scholar

141 

Fang Z, Wang G, Huang R, Liu C, Yushanjiang F, Mao T and Li J: Astilbin protects from sepsis-induced cardiac injury through the NRF2/HO-1 and TLR4/NF-κB pathway. Phytother Res. 38:1044–1058. 2024. View Article : Google Scholar

142 

Su Z, Gao M, Weng L and Xu T: Esculin targets TLR4 to protect against LPS-induced septic cardiomyopathy. Int Immunopharmacol. 131:1118972024. View Article : Google Scholar : PubMed/NCBI

143 

Shaojun Z, Yanyan X, Jian C, Xia Z, Qiang F and Saiping J: Effects of puerarin on lipopolysaccharide-induced myocardial dysfunction in isolated rat hearts. Pak J Pharm Sci. 30:1195–1202. 2017.PubMed/NCBI

144 

Xing C, Xu L and Yao Y: Beneficial role of oleuropein in sepsis-induced myocardial injury. Possible Involvement of GSK-3β/NF-κB pathway. Acta Cir Bras. 36:e3601072021. View Article : Google Scholar

145 

Shyni GL, Renjitha J, B Somappa S and Raghu KG: Zerumin A attenuates the inflammatory responses in LPS-stimulated H9c2 cardiomyoblasts. J Biochem Mol Toxicol. 35:1–11. 2021. View Article : Google Scholar : PubMed/NCBI

146 

Yan C, Kuang W, Jin L, Wang R, Niu L, Xie C, Ding J, Liao Y, Wang L, Wan H and Ma G: Carvacrol protects mice against LPS-induced sepsis and attenuates inflammatory response in macrophages by modulating the ERK1/2 pathway. Sci Rep. 13:128092023. View Article : Google Scholar : PubMed/NCBI

147 

Tang J, Hu JJ, Lu CH, Liang JN, Xiao JF, Liu YT, Lin CS and Qin ZS: Propofol inhibits lipopolysaccharide-induced tumor necrosis factor-alpha expression and myocardial depression through decreasing the generation of superoxide anion in cardiomyocytes. Oxid Med Cell Longev. 2014:1573762014. View Article : Google Scholar : PubMed/NCBI

148 

Li C, Wan W, Ye T, Sun Y, Chen X, Liu X, Shi S, Zhang Y, Qu C, Yang B, et al: Pinocembrin alleviates lipopolysaccharide-induced myocardial injury and cardiac dysfunction in rats by inhibiting p38/JNK MAPK pathway. Life Sci. 277:1194182021. View Article : Google Scholar : PubMed/NCBI

149 

Cao W, Li XQ, Zhang XN, Hou Y, Zeng AG, Xie YH and Wang SW: Madecassoside suppresses LPS-induced TNF-alpha production in cardiomyocytes through inhibition of ERK, p38, and NF-kappaB activity. Int Immunopharmacol. 10:723–729. 2010. View Article : Google Scholar : PubMed/NCBI

150 

Wang B, Chen L, Dai L, Fang W and Wang H: Alisol B 23-Acetate ameliorates Lipopolysaccharide-induced cardiac dysfunction by suppressing Toll-Like Receptor 4 (TLR4)/NADPH Oxidase 2 (NOX2) signaling pathway. Med Sci Monit. 25:8472–8481. 2019. View Article : Google Scholar : PubMed/NCBI

151 

Zhang M, Wang X, Wang X, Hou X, Teng P, Jiang Y, Zhang L, Yang X, Tian J, Li G, et al: Oxymatrine protects against myocardial injury via inhibition of JAK2/STAT3 signaling in rat septic shock. Mol Med Rep. 7:1293–1299. 2013. View Article : Google Scholar : PubMed/NCBI

152 

Zhang M, Wang X, Bai B, Zhang R, Li Y and Wang Y: Oxymatrine protects against sepsis-induced myocardial injury via inhibition of the TNF-α/p38-MAPK/caspase-3 signaling pathway. Mol Med Rep. 14:551–559. 2016. View Article : Google Scholar : PubMed/NCBI

153 

Zhao P, Wang Y, Zeng S, Lu J, Jiang TM and Li YM: Protective effect of astragaloside IV on lipopolysaccharide-induced cardiac dysfunction via downregulation of inflammatory signaling in mice. Immunopharmacol Immunotoxicol. 37:428–433. 2015. View Article : Google Scholar : PubMed/NCBI

154 

Zhai J and Guo Y: Paeoniflorin attenuates cardiac dysfunction in endotoxemic mice via the inhibition of nuclear factor-κB. Biomed Pharmacother. 80:200–206. 2016. View Article : Google Scholar : PubMed/NCBI

155 

Shang X, Lin K, Yu R, Zhu P, Zhang Y, Wang L, Xu J and Chen K: Resveratrol protects the myocardium in sepsis by activating the phosphatidylinositol 3-Kinases (PI3K)/AKT/Mammalian target of rapamycin (mTOR) pathway and inhibiting the nuclear Factor-κB (NF-κB) signaling pathway. Med Sci Monit. 25:9290–9298. 2019. View Article : Google Scholar : PubMed/NCBI

156 

Lee AS, Chen WP, Kuo YL, Ho YJ, Lee SS and Su MJ: Thaliporphine preserves cardiac function of endotoxemic rabbits by both directly and indirectly attenuating NFκB signaling pathway. PLoS One. 7:e391742012. View Article : Google Scholar

157 

He H, Chang X, Gao J, Zhu L, Miao M and Yan T: Salidroside mitigates Sepsis-Induced myocarditis in rats by regulating IGF-1/PI3K/Akt/GSK-3β signaling. Inflammation. 38:2178–2184. 2015. View Article : Google Scholar : PubMed/NCBI

158 

Li C, Hou D, Huang Y, Liu Y, Li Y and Wang C: Corylin alleviated sepsis-associated cardiac dysfunction via attenuating inflammation through downregulation of microRNA-214-5p. Toxicol Res (Camb). 13:tfae0812024. View Article : Google Scholar : PubMed/NCBI

159 

Zhang J, Liu Y and Liu L: Hyperoside prevents sepsis-associated cardiac dysfunction through regulating cardiomyocyte viability and inflammation via inhibiting miR-21. Biomed Pharmacother. 138:1115242021. View Article : Google Scholar : PubMed/NCBI

160 

Liu Y, Liu L and Zhang J: Protective role of matrine in sepsis-associated cardiac dysfunction through regulating the lncRNA PTENP1/miR-106b-5p axis. Biomed Pharmacother. 134:1111122021. View Article : Google Scholar

161 

Zhao H, Wang Y and Zhu X: Chrysophanol exerts a protective effect against sepsis-induced acute myocardial injury through modulating the microRNA-27b-3p/Peroxisomal proliferating-activated receptor gamma axis. Bioengineered. 13:12673–12690. 2022. View Article : Google Scholar : PubMed/NCBI

162 

Athapaththu A, Lee KT, Kavinda MHD, Lee S, Kang S, Lee MH, Kang CH, Choi YH and Kim GY: Pinostrobin ameliorates lipopolysaccharide (LPS)-induced inflammation and endotoxemia by inhibiting LPS binding to the TLR4/MD2 complex. Biomed Pharmacother. 156:1138742022. View Article : Google Scholar : PubMed/NCBI

163 

Feng J, Liu Z, Chen H, Zhang M, Ma X, Han Q, Lu D and Wang C: Protective effect of cynaroside on sepsis-induced multiple organ injury through Nrf2/HO-1-dependent macrophage polarization. Eur J Pharmacol. 911:1745222021. View Article : Google Scholar : PubMed/NCBI

164 

Jinzhong Wang MS and Jian Fu MS: STAT3/FoxO3a/Sirt1 pathway inhibition by ginsenoside Rc ameliorates cardiomyocyte damage in septic cardiomyopathy by altering macrophage polarization. J Mol Histol. 56:1482025. View Article : Google Scholar : PubMed/NCBI

165 

Li F, Lang F, Wang Y, Zhai C, Zhang C, Zhang L and Hao E: Cyanidin ameliorates endotoxin-induced myocardial toxicity by modulating inflammation and oxidative stress through mitochondria and other factors. Food Chem Toxicol. 120:104–111. 2018. View Article : Google Scholar : PubMed/NCBI

166 

Chen HM, Liou SF, Hsu JH, Chen TJ, Cheng TL, Chiu CC and Yeh JL: Baicalein inhibits HMGB1 release and MMP-2/-9 expression in lipopolysaccharide-induced cardiac hypertrophy. Am J Chin Med. 42:785–797. 2014. View Article : Google Scholar : PubMed/NCBI

167 

Wu W, Wang J, Wang G, Wang F, Yang Y, Liu Z, Song Q, Chen S and Chen H: Monotropein inhibits MMP9-mediated cardiac oxidative stress, inflammation, matrix degradation and apoptosis in a mouse and cell line models of septic cardiac injury. Mol Biol Rep. 52:3292025. View Article : Google Scholar : PubMed/NCBI

168 

Cao W, Zhang W, Liu J, Wang Y, Peng X, Lu D, Qi R, Wang Y and Wang H: Paeoniflorin improves survival in LPS-challenged mice through the suppression of TNF-α and IL-1β release and augmentation of IL-10 production. Int Immunopharmacol. 11:172–178. 2011. View Article : Google Scholar

169 

Liu A, Xun S, Zhou G, Zhang Y and Lin L: Honokiol alleviates sepsis-associated cardiac dysfunction via attenuating inflammation, apoptosis and oxidative stress. J Pharm Pharmacol. 75:397–406. 2023. View Article : Google Scholar : PubMed/NCBI

170 

Tang X, Xu Y, Dai X, Xing Y, Yang D, Huang Q, Li H, Lv X, Wang Y, Lu D and Wang H: The Long-term effect of dobutamine on intrinsic myocardial function and myocardial injury in septic rats with myocardial dysfunction. Shock. 56:582–592. 2021. View Article : Google Scholar : PubMed/NCBI

171 

Tsai YC, Cheng PY, Kung CW, Peng YJ, Ke TH, Wang JJ and Yen MH: Beneficial effects of magnolol in a rodent model of endotoxin shock. Eur J Pharmacol. 641:67–73. 2010. View Article : Google Scholar : PubMed/NCBI

172 

Meng ZJ, Wang C, Meng LT, Bao BH, Wu JH and Hu YQ: Sodium tanshinone IIA sulfonate attenuates cardiac dysfunction and improves survival of rats with cecal ligation and puncture-induced sepsis. Chin J Nat Med. 16:846–855. 2018.PubMed/NCBI

173 

Dörtbudak MB, Demircioğlu M and Kapucuk FS: Micromeria congesta alleviates LPS-Induced inflammation, apoptosis, oxidative stress and DNA damage in rat heart and kidneys. Vet Med Sci. 11:e702642025. View Article : Google Scholar : PubMed/NCBI

174 

Pan J, Meng L, Li R, Wang Z, Yuan W, Li Y, Chen L, Shen Q, Liu W and Zhu L: Naringenin protects against septic cardiomyopathy in mice by targeting HIF-1α. Biochem Biophys Res Commun. 704:1496132024. View Article : Google Scholar

175 

Cheng Z, Lv D, Luo M, Wang R, Guo Y, Yang X, Huang L, Li X, Li C, Shang FF, et al: Tubeimoside I protects against sepsis-induced cardiac dysfunction via SIRT3. Eur J Pharmacol. 905:1741862021. View Article : Google Scholar : PubMed/NCBI

176 

Wang Y, Yu X, Wang F, Wang Y, Wang Y, Li H, Lv X, Lu D and Wang H: Yohimbine promotes cardiac NE release and prevents LPS-induced cardiac dysfunction via blockade of presynaptic α2A-adrenergic receptor. PLoS One. 8:e636222013. View Article : Google Scholar

177 

Duzen IV, Oguz E, Yilmaz R, Taskin A, Vuruskan E, Cekici Y, Bilgel ZG, Goksuluk H, Candemir B and Sucu M: Lycopene has a protective effect on septic shock-induced cardiac injury in rats. Bratisl Lek Listy. 120:919–923. 2019.

178 

Ben-Shaul V, Lomnitski L, Nyska A, Zurovsky Y, Bergman M and Grossman S: The effect of natural antioxidants, NAO and apocynin, on oxidative stress in the rat heart following LPS challenge. Toxicol Lett. 123:1–10. 2001. View Article : Google Scholar : PubMed/NCBI

179 

Li X, Zhang Z, Zhang X, Yin Y, Yuan X, You X and Wu J: Echinacoside prevents Sepsis-induced myocardial damage via targeting SOD2. J Med Food. 27:123–133. 2024. View Article : Google Scholar

180 

Xianchu L, Lan Z, Ming L and Yanzhi M: Protective effects of rutin on lipopolysaccharide-induced heart injury in mice. J Toxicol Sci. 43:329–337. 2018. View Article : Google Scholar : PubMed/NCBI

181 

Yang C, Wu K, Li SH and You Q: Protective effect of curcumin against cardiac dysfunction in sepsis rats. Pharm Biol. 51:482–487. 2013. View Article : Google Scholar : PubMed/NCBI

182 

Sompamit K, Kukongviriyapan U, Nakmareong S, Pannangpetch P and Kukongviriyapan V: Curcumin improves vascular function and alleviates oxidative stress in non-lethal lipopolysaccharide-induced endotoxaemia in mice. Eur J Pharmacol. 616:192–199. 2009. View Article : Google Scholar : PubMed/NCBI

183 

Kukongviriyapan U, Sompamit K, Pannangpetch P, Kukongviriyapan V and Donpunha W: Preventive and therapeutic effects of quercetin on lipopolysaccharide-induced oxidative stress and vascular dysfunction in mice. Can J Physiol Pharmacol. 90:1345–1353. 2012. View Article : Google Scholar : PubMed/NCBI

184 

Hao E, Lang F, Chen Y, Zhang H, Cong X, Shen X and Su G: Resveratrol alleviates endotoxin-induced myocardial toxicity via the Nrf2 transcription factor. PLoS One. 8:e694522013. View Article : Google Scholar : PubMed/NCBI

185 

Xingyue L, Shuang L, Qiang W, Jinjuan F and Yongjian Y: Chrysin ameliorates Sepsis-induced cardiac dysfunction through upregulating Nfr2/Heme oxygenase 1 pathway. J Cardiovasc Pharmacol. 77:491–500. 2021. View Article : Google Scholar : PubMed/NCBI

186 

Tan Y, Wan HH, Sun MM, Zhang WJ, Dong M, Ge W, Ren J and Peng H: Cardamonin protects against lipopolysaccharide-induced myocardial contractile dysfunction in mice through Nrf2-regulated mechanism. Acta Pharmacol Sin. 42:404–413. 2021. View Article : Google Scholar :

187 

Li Y, Zhang L, Zhang P and Hao Z: Dehydrocorydaline protects against Sepsis-induced myocardial injury through modulating the TRAF6/NF-κB pathway. Front Pharmacol. 12:7096042021. View Article : Google Scholar

188 

Lee YM, Cheng PY, Chim LS, Kung CW, Ka SM, Chung MT and Sheu JR: Baicalein, an active component of Scutellaria baicalensis Georgi, improves cardiac contractile function in endotoxaemic rats via induction of heme oxygenase-1 and suppression of inflammatory responses. J Ethnopharmacol. 135:179–185. 2011. View Article : Google Scholar : PubMed/NCBI

189 

Cheng PY, Lee YM, Wu YS, Chang TW, Jin JS and Yen MH: Protective effect of baicalein against endotoxic shock in rats in vivo and in vitro. Biochem Pharmacol. 73:793–804. 2007. View Article : Google Scholar

190 

Chen WP, Tzeng HJ, Ku HC, Ho YJ, Lee SS and Su MJ: Thaliporphine ameliorates cardiac depression in endotoxemic rats through attenuating TLR4 signaling in the downstream of TAK-1 phosphorylation and NF-κB signaling. Naunyn Schmiedebergs Arch Pharmacol. 382:441–453. 2010. View Article : Google Scholar : PubMed/NCBI

191 

Chen RC, Wang J, Yang L, Sun GB and Sun XB: Protective effects of ginsenoside Re on lipopolysaccharide-induced cardiac dysfunction in mice. Food Funct. 7:2278–2287. 2016. View Article : Google Scholar : PubMed/NCBI

192 

Chen L, Liu P, Feng X and Ma C: Salidroside suppressing LPS-induced myocardial injury by inhibiting ROS-mediated PI3K/Akt/mTOR pathway in vitro and in vivo. J Cell Mol Med. 21:3178–3189. 2017. View Article : Google Scholar : PubMed/NCBI

193 

Xie L, Zhao M, Zong L and Yue Y: Propofol ameliorates Sepsis-induced myocardial dysfunction via Anti-Apoptotic, Anti-Oxidative properties, and mTOR signaling. Discov Med. 36:2088–2097. 2024. View Article : Google Scholar : PubMed/NCBI

194 

Wang L, Zhao Y, Su Z, Zhao K, Li P and Xu T: Ginkgolide A targets forkhead box O1 to protect against lipopolysaccharide-induced septic cardiomyopathy. Phytother Res. 37:3309–3322. 2023. View Article : Google Scholar : PubMed/NCBI

195 

Huang L, Zheng M, Zhou Y, Zhu J, Zhu M, Zhao F and Cui S: Tanshinone IIA attenuates cardiac dysfunction in endotoxin-induced septic mice via inhibition of NADPH oxidase 2-related signaling pathway. Int Immunopharmacol. 28:444–449. 2015. View Article : Google Scholar : PubMed/NCBI

196 

Hou D, Liao H, Hao S, Liu R, Huang H and Duan C: Curcumin simultaneously improves mitochondrial dynamics and myocardial cell bioenergy after sepsis via the SIRT1-DRP1/PGC-1α pathway. Heliyon. 10:e285012024. View Article : Google Scholar

197 

Smeding L, Leong-Poi H, Hu P, Shan Y, Haitsma JJ, Horvath E, Furmli S, Masoom H, Kuiper JW, Slutsky AS, et al: Salutary effect of resveratrol on sepsis-induced myocardial depression. Crit Care Med. 40:1896–1907. 2012. View Article : Google Scholar : PubMed/NCBI

198 

Peng K, Yang F, Qiu C, Yang Y and Lan C: Rosmarinic acid protects against lipopolysaccharide-induced cardiac dysfunction via activating Sirt1/PGC-1α pathway to alleviate mitochondrial impairment. Clin Exp Pharmacol Physiol. 50:218–227. 2023. View Article : Google Scholar

199 

Li Y, Feng YF, Liu XT, Li YC, Zhu HM, Sun MR, Li P, Liu B and Yang H: Songorine promotes cardiac mitochondrial biogenesis via Nrf2 induction during sepsis. Redox Biol. 38:1017712021. View Article : Google Scholar

200 

Yang Z, Liu Y, Deng W, Dai J, Li F, Yuan Y, Wu Q, Zhou H, Bian Z and Tang Q: Hesperetin attenuates mitochondria-dependent apoptosis in lipopolysaccharide-induced H9C2 cardiomyocytes. Mol Med Rep. 9:1941–1946. 2014. View Article : Google Scholar : PubMed/NCBI

201 

Yang YP, Zhao JQ, Gao HB, Li JJ, Li XL, Niu XL, Lei YH and Li X: Tannic acid alleviates lipopolysaccharide-induced H9C2 cell apoptosis by suppressing reactive oxygen species-mediated endoplasmic reticulum stress. Mol Med Rep. 24:5352021. View Article : Google Scholar :

202 

Xie WJ, Liu M, Zhang X, Zhang YG, Jian ZH and Xiong XX: Astaxanthin suppresses LPS-induced myocardial apoptosis by regulating PTP1B/JNK pathway in vitro. Int Immunopharmacol. 127:1113952024. View Article : Google Scholar

203 

Ye G, Wang M, Liu D, Cheng L, Yin X, Zhang Q and Liu W: Mechanism of naringenin blocking the protection of LTB4/BLT1 receptor against septic cardiac dysfunction. Ann Clin Lab Sci. 50:769–774. 2020.PubMed/NCBI

204 

Zhao H, Chen Y, Qian L, Du L, Wu X, Tian Y, Deng C, Liu S, Yang W, Lu C, et al: Lycorine protects against septic myocardial injury by activating AMPK-related pathways. Free Radic Biol Med. 197:1–14. 2023. View Article : Google Scholar : PubMed/NCBI

205 

Miao H, Tang X, Cui Y, Shi J, Xiong X, Wang C and Zhang Y: Obeticholic acid inhibit mitochondria dysfunction via regulating ERK1/2-DRP pathway to exert protective effect on lipopolysaccharide-induced myocardial injury. Adv Biol (Weinh). 8:e23005762024. View Article : Google Scholar : PubMed/NCBI

206 

Qi Z, Wang R, Liao R, Xue S and Wang Y: Neferine ameliorates Sepsis-induced myocardial dysfunction through Anti-apoptotic and antioxidative effects by regulating the PI3K/AKT/mTOR signaling pathway. Front Pharmacol. 12:7062512021. View Article : Google Scholar : PubMed/NCBI

207 

Liu Z, Pan H, Zhang Y, Zheng Z, Xiao W, Hong X, Chen F, Peng X, Pei Y, Rong J, et al: Ginsenoside-Rg1 attenuates sepsis-induced cardiac dysfunction by modulating mitochondrial damage via the P2X7 receptor-mediated Akt/GSK-3β signaling pathway. J Biochem Mol Toxicol. 36:e228852022. View Article : Google Scholar

208 

Zhu X, Sun M, Guo H, Lu G, Gu J, Zhang L, Shi L, Gao J, Zhang D, Wang W, et al: Verbascoside protects from LPS-induced septic cardiomyopathy via alleviating cardiac inflammation, oxidative stress and regulating mitochondrial dynamics. Ecotoxicol Environ Saf. 233:1133272022. View Article : Google Scholar : PubMed/NCBI

209 

Lu C, Lei W, Sun M, Wu X, Liu Q, Liu J, Yang Y, Yang W, Zhang Z, Li X, et al: Identification of CCR2 as a hub in septic myocardial injury and cardioprotection of silibinin. Free Radic Biol Med. 197:46–57. 2023. View Article : Google Scholar : PubMed/NCBI

210 

Sun M, Zhao H, Jin Z, Lei W, Deng C, Yang W, Lu C, Hou Y, Zhang Y, Tang R, et al: Silibinin protects against sepsis and septic myocardial injury in an NR1H3-dependent pathway. Free Radic Biol Med. 187:141–157. 2022. View Article : Google Scholar : PubMed/NCBI

211 

Sun HJ, Zheng GL, Wang ZC, Liu Y, Bao N, Xiao PX, Lu QB and Zhang JR: Chicoric acid ameliorates sepsis-induced cardiomyopathy via regulating macrophage metabolism reprogramming. Phytomedicine. 123:1551752024. View Article : Google Scholar

212 

Chen M, Huang S, Weng S, Weng J, Guo R, Shi B and Liu D: Songorine ameliorates LPS-induced sepsis cardiomyopathy by Wnt/β-catenin signaling pathway-mediated mitochondrial biosynthesis. Naunyn Schmiedebergs Arch Pharmacol. 397:4713–4725. 2024. View Article : Google Scholar

213 

Hwang HR, Tai BY, Cheng PY, Chen PN, Sung PJ, Wen ZH and Hsu CH: Excavatolide B modulates the electrophysiological characteristics and calcium homeostasis of atrial myocytes. Mar Drugs. 15:252017. View Article : Google Scholar : PubMed/NCBI

214 

Chen R, Zheng A, Wang Y, Guo L, Dou H, Lu L, Rafiq M, Li P, Chen X and Xiao Q: Salvianolic acid B improves mitochondrial dysfunction of septic cardiomyopathy via enhancing ATF5-mediated mitochondrial unfolded protein response. Toxicol Appl Pharmacol. 491:1170722024. View Article : Google Scholar : PubMed/NCBI

215 

Nong Y, Lu J, Yu D and Wei X: Neohesperidin dihydrochalcone alleviates Lipopolysaccharide-induced vascular endothelium dysfunction by regulating antioxidant capacity. Immun Inflamm Dis. 12:e701072024. View Article : Google Scholar : PubMed/NCBI

216 

Tang F, Liu D, Wan F, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H and Peng C: Ameliorative effect of anisodamine (654-1/654-2) against myocardial dysfunction induced by septic shock via the NF-κB/NLRP-3 or the PI3K-AKT/NF-κB pathway. Phytomedicine. 123:1552772024. View Article : Google Scholar

217 

Chiorcea-Paquim AM: Electrochemistry of flavonoids: A comprehensive review. Int J Mol Sci. 24:156672023. View Article : Google Scholar : PubMed/NCBI

218 

Billowria K, Ali R, Rangra NK, Kumar R and Chawla PA: Bioactive flavonoids: A comprehensive review on pharmacokinetics and analytical aspects. Crit Rev Anal Chem. 54:1002–1016. 2024. View Article : Google Scholar

219 

Jomova K, Alomar SY, Valko R, Liska J, Nepovimova E, Kuca K and Valko M: Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem Biol Interact. 413:1114892025. View Article : Google Scholar : PubMed/NCBI

220 

Patel S: Plant-derived cardiac glycosides: Role in heart ailments and cancer management. Biomed Pharmacother. 84:1036–1041. 2016. View Article : Google Scholar : PubMed/NCBI

221 

de Araújo FF, de Paulo Farias D, Neri-Numa IA and Pastore GM: Polyphenols and their applications: An approach in food chemistry and innovation potential. Food Chem. 338:1275352021. View Article : Google Scholar

222 

Cinelli MA and Jones AD: Alkaloids of the genus datura: Review of a rich resource for natural product discovery. Molecules. 26:26292021. View Article : Google Scholar : PubMed/NCBI

223 

Jolly A, Hour Y and Lee YC: An outlook on the versatility of plant saponins: A review. Fitoterapia. 174:1058582024. View Article : Google Scholar : PubMed/NCBI

224 

Huang S, Liu D, Sun J, Zhang H, Zhang J, Wang Q, Gan L, Qu G, Qiu J, Deng J, et al: Tim-3 regulates sepsis-induced immunosuppression by inhibiting the NF-κB signaling pathway in CD4 T cells. Mol Ther. 30:1227–1238. 2022. View Article : Google Scholar

225 

Chen XS, Wang SH, Liu CY, Gao YL, Meng XL, Wei W, Shou ST, Liu YC and Chai YF: Losartan attenuates sepsis-induced cardiomyopathy by regulating macrophage polarization via TLR4-mediated NF-κB and MAPK signaling. Pharmacol Res. 185:1064732022. View Article : Google Scholar

226 

Yang Q, Wang Y, Cao G, Li X and Zhao T: Anti-sepsis effect of Xiaochaihu decoction based on the TLR4/MyD88/NF-κB signalling pathway. Heliyon. 10:e267122024. View Article : Google Scholar

227 

Huang L, Li Y, Cheng Z, Lv Z, Luo S and Xia Y: PCSK9 promotes endothelial dysfunction during sepsis via the TLR4/MyD88/NF-κB and NLRP3 Pathways. Inflammation. 46:115–128. 2023. View Article : Google Scholar

228 

Wu Y, Wang Q, Li M, Lao J, Tang H, Ming S, Wu M, Gong S, Li L, Liu L and Huang X: SLAMF7 regulates the inflammatory response in macrophages during polymicrobial sepsis. J Clin Invest. 133:e1502242023. View Article : Google Scholar : PubMed/NCBI

229 

Al-Kadi A, Anter AF, Rofaeil RR, Sayed-Ahmed MM, Hafez S and Ahmed AF: Endothelin system blockade extenuates Sepsis-induced acute heart and kidney injuries via modulating ET-1/Klotho/p38-MAPK. Clin Exp Pharmacol Physiol. 52:e700422025. View Article : Google Scholar : PubMed/NCBI

230 

Wang Y, Yu W, Shi C and Hu P: Crocetin attenuates Sepsis-induced cardiac dysfunction via regulation of inflammatory response and mitochondrial function. Front Physiol. 11:5142020. View Article : Google Scholar : PubMed/NCBI

231 

Li J, Wang L, Wang B, Zhang Z, Jiang L, Qin Z, Zhao Y and Su B: NOX4 is a potential therapeutic target in septic acute kidney injury by inhibiting mitochondrial dysfunction and inflammation. Theranostics. 13:2863–2878. 2023. View Article : Google Scholar : PubMed/NCBI

232 

Pan T, Sun S, Chen Y, Tian R, Chen E, Tan R, Wang X, Liu Z, Liu J and Qu H: Immune effects of PI3K/Akt/HIF-1α-regulated glycolysis in polymorphonuclear neutrophils during sepsis. Crit Care. 26:292022. View Article : Google Scholar

233 

Ma L, Zhang R, Li D, Qiao T and Guo X: Fluoride regulates chondrocyte proliferation and autophagy via PI3K/AKT/mTOR signaling pathway. Chem Biol Interact. 349:1096592021. View Article : Google Scholar : PubMed/NCBI

234 

Lee J, Kim J, Lee JH, Choi YM, Choi H, Cho HD, Cha GH, Lee YH, Jo EK, Park BH and Yuk JM: SIRT1 promotes host protective immunity against toxoplasma gondii by controlling the FoxO-autophagy axis via the AMPK and PI3K/AKT signalling pathways. Int J Mol Sci. 23:135782022. View Article : Google Scholar : PubMed/NCBI

235 

Yin Z, Tian L, Kou W, Cao G, Wang L, Xia Y, Lin Y, Tang S, Zhang J and Yang H: Xiyangshen Sanqi Danshen granules attenuated D-gal-induced C57BL/6J mouse aging through the AMPK/SIRT1 signaling pathway. Phytomedicine. 136:1562132025. View Article : Google Scholar

236 

Chen Y, Chen J, Xing Z, Peng C and Li D: Autophagy in neuroinflammation: A focus on epigenetic regulation. Aging Dis. 15:739–754. 2024. View Article : Google Scholar :

237 

Lu SM, Yang B, Tan ZB, Wang HJ, Xie JD, Xie MT, Jiang WH, Huang JZ, Li J, Zhang L, et al: TaoHe ChengQi decoction ameliorates sepsis-induced cardiac dysfunction through anti-ferroptosis via the Nrf2 pathway. Phytomedicine. 129:1555972024. View Article : Google Scholar : PubMed/NCBI

238 

Tang F, Yan YM, Yan HL, Wang LX, Hu CJ, Wang HL, Ao H, Peng C and Tan YZ: Chuanxiongdiolides R4 and R5, phthalide dimers with a complex polycyclic skeleton from the aerial parts of Ligusticum chuanxiong and their vasodilator activity. Bioorg Chem. 107:1045232021. View Article : Google Scholar

239 

Zhang T, Lu M, Yang Y, Ji X, Gu H, Sun Y, Chen C and Sun T: Cold-adapted nanozymes. Adv Healthc Mater. 14:e25012112025. View Article : Google Scholar : PubMed/NCBI

240 

Li S, Wang F, Hao L, Zhang P, Song G, Zhang Y, Wang C, Wang Z and Wu Q: Enhancing peroxidase activity of NiCo2O4 nanoenzyme by Mn doping for catalysis of CRISPR/Cas13a-mediated non-coding RNA detection. Int J Biol Macromol. 283:1375942024. View Article : Google Scholar

241 

Meng X, Fan K and Yan X: Nanozymes: An emerging field bridging nanotechnology and enzymology. Sci China Life Sci. 62:1543–1546. 2019. View Article : Google Scholar : PubMed/NCBI

242 

Cao X, Jiang H, Huang X, Sun D and Qi G: Hydrogel patch doped with nanoenzyme for SERS detection of hydrogen peroxide in complex body fluids. Talanta. 285:1273282025. View Article : Google Scholar

243 

Jiang C, Shi Q, Yang J, Ren H, Zhang L, Chen S, Si J, Liu Y, Sha D, Xu B and Ni J: Ceria nanozyme coordination with curcumin for treatment of sepsis-induced cardiac injury by inhibiting ferroptosis and inflammation. J Adv Res. 63:159–170. 2024. View Article : Google Scholar :

244 

Li S, Wang K, Jiang K, Xing D, Deng R, Xu Y, Ding Y, Guan H, Chen LL, Wang D, et al: Brazilin-Ce nanoparticles attenuate inflammation by de/anti-phosphorylation of IKKβ. Biomaterials. 305:1224662024. View Article : Google Scholar

245 

Mai BT, Fernandes S, Balakrishnan PB and Pellegrino T: Nanosystems based on magnetic nanoparticles and thermo\or pH-Responsive polymers: An update and future perspectives. Acc Chem Res. 51:999–1013. 2018. View Article : Google Scholar : PubMed/NCBI

246 

De Jong WH and Borm PJ: Drug delivery and nanoparticles: Applications and hazards. Int J Nanomedicine. 3:133–149. 2008. View Article : Google Scholar :

247 

Wang J, Wang H, Zhu R, Liu Q, Fei J and Wang S: Anti-inflammatory activity of Curcumin-loaded solid lipid nanoparticles in IL-1β transgenic mice subjected to the lipopolysaccharide-induced sepsis. Biomaterials. 53:475–483. 2015. View Article : Google Scholar

248 

Rattis BAC, Piva HL, Duarte A, Gomes FGFLR, Lellis JR, Soave DF, Ramos SG, Tedesco AC and Celes MRN: Modulation of the mTOR pathway by curcumin in the heart of septic mice. Pharmaceutics. 14:22772022. View Article : Google Scholar : PubMed/NCBI

249 

Tian B, Hua S and Liu J: Cyclodextrin-based delivery systems for chemotherapeutic anticancer drugs: A review. Carbohydr Polym. 232:1158052020. View Article : Google Scholar : PubMed/NCBI

250 

Sahu KM, Patra S and Swain SK: Host-guest drug delivery by β-cyclodextrin assisted polysaccharide vehicles: A review. Int J Biol Macromol. 240:1243382023. View Article : Google Scholar

251 

Heimfarth L, Dos Santos KS, Monteiro BS, de Souza Oliveira AK, Coutinho HDM, Menezes IRA, Dos Santos MRV, de Souza Araújo AA, Picot L, de Oliveira Júnior RG, et al: The protective effects of naringenin, a citrus flavonoid, non-complexed or complexed with hydroxypropyl-β-cyclodextrin against multiorgan damage caused by neonatal endotoxemia. Int J Biol Macromol. 264:1305002024. View Article : Google Scholar

252 

Penalva R, González-Navarro CJ, Gamazo C, Esparza I and Irache JM: Zein nanoparticles for oral delivery of quercetin: Pharmacokinetic studies and preventive anti-inflammatory effects in a mouse model of endotoxemia. Nanomedicine. 13:103–110. 2017. View Article : Google Scholar

253 

Wang S, Tan KS, Beng H, Liu F, Huang J, Kuai Y, Zhang R and Tan W: Protective effect of isosteviol sodium against LPS-induced multiple organ injury by regulating of glycerophospholipid metabolism and reducing macrophage-driven inflammation. Pharmacol Res. 172:1057812021. View Article : Google Scholar : PubMed/NCBI

254 

Liu S, Yao C, Xie J, Liu H, Wang H, Lin Z, Qin B, Wang D, Lu W, Ma X, et al: Effect of an Herbal-based injection on 28-Day mortality in patients with sepsis: The EXIT-SEP randomized clinical trial. JAMA Intern Med. 183:647–655. 2023. View Article : Google Scholar : PubMed/NCBI

255 

Zhang H, Wei L, Zhao G, Liu S, Zhang Z, Zhang J and Yang Y: Protective effect of Xuebijing injection on myocardial injury in patients with sepsis: A randomized clinical trial. J Tradit Chin Med. 36:706–710. 2016. View Article : Google Scholar : PubMed/NCBI

256 

Wu X, He C, Liu C, Xu X, Chen C, Yang H, Shi H, Fei Y, Sun Y, Zhou S and Fang B: Mechanisms of JinHong Formula on treating sepsis explored by randomized controlled trial combined with network pharmacology. J Ethnopharmacol. 305:1160402023. View Article : Google Scholar

257 

Huang N, Tam YH, Zhang Z, Kao X, Yang Z, Xu W, Yuan K, He M and Chen J: Efficacy and safety of Dachaihu decoction for sepsis: A randomized controlled trial. Phytomedicine. 136:1563112025. View Article : Google Scholar

258 

Liao J, Qin C, Wang Z, Gao L, Zhang S, Feng Y, Liu J and Tao L: Effect of shenfu injection in patients with septic shock: A systemic review and meta-analysis for randomized clinical trials. J Ethnopharmacol. 320:1174312024. View Article : Google Scholar

259 

Yu Y, Zhu C, Hong Y, Chen L, Huang Z, Zhou J, Tian X, Liu D, Ren B, Zhang C, et al: Effectiveness of anisodamine for the treatment of critically ill patients with septic shock: A multicentre randomized controlled trial. Crit Care. 25:3492021. View Article : Google Scholar : PubMed/NCBI

260 

Zhang F, Mei X, Zhou P, Tian YP, Liu JX, Dong X, Yuan DS, Lin ZF, Zhang L, Lin JH, et al: Anisodamine hydrobromide in the treatment of critically ill patients with septic shock: A multi-center randomized controlled trial. Ann Med. 55:22643182023. View Article : Google Scholar

261 

Guo X, Luo W, Wu L, Zhang L, Chen Y, Li T, Li H, Zhang W, Liu Y, Zheng J and Wang Y: Natural products from herbal medicine Self-Assemble into advanced bioactive materials. Adv Sci (Weinh). 11:e24033882024. View Article : Google Scholar : PubMed/NCBI

262 

Li J, Sun S, Zhu D, Mei X, Lyu Y, Huang K, Li Y, Liu S, Wang Z, Hu S, et al: Inhalable stem cell exosomes promote heart repair after myocardial infarction. Circulation. 150:710–723. 2024. View Article : Google Scholar : PubMed/NCBI

263 

Yuan Y, Mei Z, Qu Z, Li G, Yu S, Liu Y, Liu K, Shen Z, Pu J, Wang Y, et al: Exosomes secreted from cardiomyocytes suppress the sensitivity of tumor ferroptosis in ischemic heart failure. Signal Transduct Target Ther. 8:1212023. View Article : Google Scholar : PubMed/NCBI

264 

Tirziu D, Giordano FJ and Simons M: Cell communications in the heart. Circulation. 122:928–937. 2010. View Article : Google Scholar : PubMed/NCBI

265 

Hu Y, Chen H, Zhang L, Lin X, Li X, Zhuang H, Fan H, Meng T, He Z, Huang H, et al: The AMPK-MFN2 axis regulates MAM dynamics and autophagy induced by energy stresses. Autophagy. 17:1142–1156. 2021. View Article : Google Scholar :

266 

Wu S, Lu Q, Ding Y, Wu Y, Qiu Y, Wang P, Mao X, Huang K, Xie Z and Zou MH: Hyperglycemia-driven inhibition of AMP-activated protein kinase α2 induces diabetic cardiomyopathy by promoting mitochondria-associated endoplasmic reticulum membranes in vivo. Circulation. 139:1913–1936. 2019. View Article : Google Scholar : PubMed/NCBI

267 

Zhou ZK, Yu MM, Shou ST, Chai YF and Liu YC: Interaction between gut-heart axis in sepsis-induced cardiomyopathy. Pharmacol Res. 217:1078062025. View Article : Google Scholar : PubMed/NCBI

268 

Yuzefpolskaya M, Bohn B, Nasiri M, Zuver AM, Onat DD, Royzman EA, Nwokocha J, Mabasa M, Pinsino A, Brunjes D, et al: Gut microbiota, endotoxemia, inflammation, and oxidative stress in patients with heart failure, left ventricular assist device, and transplant. J Heart Lung Transplant. 39:880–890. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Tang F, Liu D, Zhu S, Zhou H and Qiu X: <p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>. Int J Mol Med 57: 71, 2026.
APA
Tang, F., Liu, D., Zhu, S., Zhou, H., & Qiu, X. (2026). <p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>. International Journal of Molecular Medicine, 57, 71. https://doi.org/10.3892/ijmm.2026.5742
MLA
Tang, F., Liu, D., Zhu, S., Zhou, H., Qiu, X."<p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>". International Journal of Molecular Medicine 57.3 (2026): 71.
Chicago
Tang, F., Liu, D., Zhu, S., Zhou, H., Qiu, X."<p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>". International Journal of Molecular Medicine 57, no. 3 (2026): 71. https://doi.org/10.3892/ijmm.2026.5742
Copy and paste a formatted citation
x
Spandidos Publications style
Tang F, Liu D, Zhu S, Zhou H and Qiu X: <p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>. Int J Mol Med 57: 71, 2026.
APA
Tang, F., Liu, D., Zhu, S., Zhou, H., & Qiu, X. (2026). <p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>. International Journal of Molecular Medicine, 57, 71. https://doi.org/10.3892/ijmm.2026.5742
MLA
Tang, F., Liu, D., Zhu, S., Zhou, H., Qiu, X."<p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>". International Journal of Molecular Medicine 57.3 (2026): 71.
Chicago
Tang, F., Liu, D., Zhu, S., Zhou, H., Qiu, X."<p>Natural products as multi‑target therapies for sepsis‑induced myocardial dysfunction (Review)</p>". International Journal of Molecular Medicine 57, no. 3 (2026): 71. https://doi.org/10.3892/ijmm.2026.5742
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team