1
|
Stevens DL, Bryant AE. Complexities of cardiomyopathy in septic shock. Curr Opin Infect Dis 2025; 38:214-221. [PMID: 40127058 DOI: 10.1097/qco.0000000000001102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
PURPOSE OF REVIEW This review highlights the complex pathophysiology of myocardial dysfunction in septic shock and emphasizes the need for early and repeated hemodynamic assessments to improve outcome. RECENT FINDINGS Septic cardiomyopathy is a complex, dynamic process driven by multiple mechanisms such as direct myocardial depression induced by host immune mediators (e.g., cytokines, nitric oxide) and/or bacterial toxins, and mitochondrial metabolic dysfunction. Recent echocardiography studies have described multiple unique hemodynamic clusters (phenotypes) that correlated with clinical outcomes. Similarly, serial echocardiography findings and mean arterial pressure abnormalities in patients with Streptococcal Toxic Shock Syndrome (StrepTSS) yielded three distinct hemodynamic groups that predicted mortality and morbidity. Because excessive use of fluids and/or vasopressors can be detrimental, especially in patients with microvascular injury or cardiomyopathy, application of the cardiovascular performance criteria of these different phenotypes could better inform management decisions in real time and improve outcome. SUMMARY Septic cardiomyopathy is a dynamic, multidimensional response of the myocardium to infection involving both normal and dysregulated immune responses in which the measurable changes in myocardial function predict outcomes. This current paradigm mandates that functional parameters of cardiac performance be measured early and repeatedly throughout the disease course using echocardiography to guide treatment and improve outcome.
Collapse
Affiliation(s)
- Dennis L Stevens
- University of Washington School of Medicine, Seattle, Washington
| | | |
Collapse
|
2
|
Habib T, Ahmed I, Abayazeed R, Montasser M. The prognostic role of tricuspid annular plane systolic excursion in critically ill patients with septic shock. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2025; 5:24. [PMID: 40307938 PMCID: PMC12042442 DOI: 10.1186/s44158-025-00227-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/22/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION The right ventricle (RV) may play a crucial role in predicting prognosis in critical settings. The value of the tricuspid annular plane systolic excursion (TAPSE) has been shown in the prognosis of cardiac patients, such as those with heart failure and pulmonary hypertension. The aim of this study was to evaluate the possible prognostic performance of RV dysfunction, as assessed by the TAPSE, in noncardiac septic shock patients. METHODOLOGY One hundred critically ill adult patients diagnosed with septic shock were enrolled directly after admission. The TAPSE was measured within 24 h. Patients were analyzed according to 28-day mortality and divided into non-survivors and survivors. RESULTS The overall 28-day mortality rate was 62%. TAPSE showed a strong negative correlation with APACHE-II (r = - 0.569, p < 0.001) and moderately negatively correlated with the SOFA score (r = - 0.448, p = 0.001). TAPSE (at a cutoff point of 2 cm) was a very good tool (area under curve = 0.887) for predicting 28-day mortality (95% confidence interval CI 0.770-0.980, p < 0.0001). CONCLUSION Early echocardiographic assessment of RV dysfunction to measure TAPSE might be of prognostic importance in noncardiac patients with septic shock, as a TAPSE less than 2 cm was useful for predicting poor outcomes. TRIAL REGISTRATION clinicaltrials.gov, NCT06008067. Registered 18 July 2023 registered. TAPSESEPTIC study.
Collapse
Affiliation(s)
- Tamer Habib
- Critical Care Medicine Department, Faculty of Medicine, Alexandria University, Alexandria, 21111, Egypt.
| | - Islam Ahmed
- Public Health and Community Medicine Department, Faculty of Medicine, Suez-Canal University, Ismailia, Egypt
- Pharmacy Practice and Clinical Pharmacy Department, Faculty of Pharmacy, King Salman International University, South-Sinai, El Tor, Egypt
| | - Rasha Abayazeed
- Cardiology and Angiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mina Montasser
- Emergency Medicine Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
3
|
Mai A, Yasarlar Y, Awad M, Terasaki Y, Zehr K, Soubani AO, Esber Z. The Role of Venoarterial Ecmo in the Management of Sepsis-Induced Cardiomyopathy and Refractory Shock: An Analytic Review. J Intensive Care Med 2025:8850666251321788. [PMID: 40255084 DOI: 10.1177/08850666251321788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Sepsis-induced cardiomyopathy is a severe complication seen in patients with refractory septic shock, characterized by cardiac dysfunction due to an overwhelming inflammatory response secondary to infection. Despite its reversible nature, sepsis-induced cardiomyopathy (SIC) can significantly increase mortality in septic shock patients despite treating the underlying infection. Prompt recognition and aggressive intervention can potentially support the recovery of cellular injuries caused by inflammation and improve outcomes. Recently, venoarterial-extracorporeal membrane oxygenation (VA-ECMO) has gained attention as a potential mechanical support for managing these patients with SIC and cardiogenic shock. However, the lack of consensus in defining SIC and the variation in inclusion criteria for VA-ECMO implementation in acute sepsis-induced cardiogenic shock made it challenging to interpret the results of related studies. A more specific definition of SIC and careful selection of patients who may benefit from ECMO support could improve outcomes in this population. This analytic review discusses the mechanisms leading to cardiomyocyte injury and SIC, and summarizes recent studies including the efficacy of VA-ECMO in managing refractory septic shock due to SIC.
Collapse
Affiliation(s)
- AnhTuan Mai
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, DMC Sinai-Grace Hospital, Detroit, Michigan, USA
| | - Yusuf Yasarlar
- Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mohammed Awad
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yusuke Terasaki
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Michael & Marian Ilitch Department of Surgery, Division of Cardiothoracic Surgery, Detroit, Michigan, USA
| | - Kenton Zehr
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Michael & Marian Ilitch Department of Surgery, Division of Cardiothoracic Surgery, Detroit, Michigan, USA
| | - Ayman O Soubani
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Zahia Esber
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
4
|
Piccioni A, Baroni S, Rozzi G, Belvederi F, Leggeri S, Spagnuolo F, Novelli M, Pignataro G, Candelli M, Covino M, Gasbarrini A, Franceschi F. Evaluation of Presepsin for Early Diagnosis of Sepsis in the Emergency Department. J Clin Med 2025; 14:2480. [PMID: 40217929 PMCID: PMC11989492 DOI: 10.3390/jcm14072480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Background: to date, there are no specific markers available for diagnosing sepsis. Diagnosis is, indeed, mainly determined by clinical suspicion and the evaluation of the patient's overall condition. This evaluation involves assessing various inflammatory markers, such as C-reactive protein (CRP) and procalcitonin (PCT), along with markers of tissue hypoxia, such as serum lactate. Additionally, it includes scores that account for complete blood count (CBC), organ function markers, and the patient's vital parameters, including SOFA, qSOFA, SIRS, and NEWS. Over the years, various potential biomarkers have been studied; among these presepsin appears to offer some significant advantages. Objective: Presepsin, which is the N-terminal fragment of the soluble component of CD14, is primarily elevated in infectious conditions. Its levels rise much earlier in the context of infection compared to currently used biomarkers. As a result, Presepsin shows promise for the early identification of septic patients and could aid in prognostic assessment, allowing clinicians to prioritize care for critically ill individuals. Methods: this study aims to evaluate the role of serum presepsin in the early diagnosis of sepsis in patients who present to the emergency room with a clinical suspicion of sepsis. The secondary objectives include comparing the diagnostic performance of presepsin with traditional biomarkers currently used for sepsis diagnosis and assessing its utility as a prognostic biomarker for mortality risk stratification, in comparison with validated severity prediction scores. Result: Presepsin had valuable diagnostic utility for sepsis (AUC 0.946, p < 0.001) comparable to PCT (AUC 0.905, p < 0.001). Conclusions: the combination of Presepsin, PCT, and EWS yielded the highest diagnostic accuracy for sepsis.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (G.P.); (M.C.); (M.C.); (F.F.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| | - Silvia Baroni
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Laboratory and Hematological Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (S.L.)
| | - Gloria Rozzi
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| | - Fabio Belvederi
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Laboratory and Hematological Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (S.L.)
| | - Simone Leggeri
- Unit of Chemistry, Biochemistry and Clinical Molecular Biology, Department of Laboratory and Hematological Sciences, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.B.); (S.L.)
| | - Fabio Spagnuolo
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| | - Michela Novelli
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| | - Giulia Pignataro
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (G.P.); (M.C.); (M.C.); (F.F.)
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (G.P.); (M.C.); (M.C.); (F.F.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (G.P.); (M.C.); (M.C.); (F.F.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| | - Antonio Gasbarrini
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy; (G.P.); (M.C.); (M.C.); (F.F.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (S.B.); (G.R.); (F.S.); (M.N.); (A.G.)
| |
Collapse
|
5
|
Malmström E, Malmström L, Hauri S, Mohanty T, Scott A, Karlsson C, Gueto-Tettay C, Åhrman E, Nozohoor S, Tingstedt B, Regner S, Elfving P, Bjermer L, Forsvall A, Doyle A, Magnusson M, Hedenfalk I, Kannisto P, Brandt C, Nilsson E, Dahlin LB, Malm J, Linder A, Niméus E, Malmström J. Human proteome distribution atlas for tissue-specific plasma proteome dynamics. Cell 2025:S0092-8674(25)00286-7. [PMID: 40203824 DOI: 10.1016/j.cell.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/16/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025]
Abstract
The plasma proteome is maintained by the influx and efflux of proteins from surrounding organs and cells. To quantify the extent to which different organs and cells impact the plasma proteome in healthy and diseased conditions, we developed a mass-spectrometry-based proteomics strategy to infer the tissue origin of proteins detected in human plasma. We first constructed an extensive human proteome atlas from 18 vascularized organs and the 8 most abundant cell types in blood. The atlas was interfaced with previous RNA and protein atlases to objectively define proteome-wide protein-organ associations to infer the origin and enable the reproducible quantification of organ-specific proteins in plasma. We demonstrate that the resource can determine disease-specific quantitative changes of organ-enriched protein panels in six separate patient cohorts, including sepsis, pancreatitis, and myocardial injury. The strategy can be extended to other diseases to advance our understanding of the processes contributing to plasma proteome dynamics.
Collapse
Affiliation(s)
- Erik Malmström
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden; Emergency Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Skåne University Hospital, Lund, Sweden; Department of Emergency medicine and Internal medicine, Emergency department, Skåne University Hospital, Lund, Sweden
| | - Lars Malmström
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Simon Hauri
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden; Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Tirthankar Mohanty
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Aaron Scott
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Christofer Karlsson
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Carlos Gueto-Tettay
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Emma Åhrman
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Shahab Nozohoor
- Department of Cardiothoracic and Vascular Surgery, Lund University and Skane University Hospital, Lund, Sweden
| | - Bobby Tingstedt
- Division of Surgery, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Sara Regner
- Department of Clinical Sciences Malmö, Section for Surgery, Lund University, 214 28 Malmö, Sweden; Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Peter Elfving
- Division of Urology, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory Medicine & Allergology, Skåne University Hospital, Lund, Sweden
| | - Andreas Forsvall
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden; Department of Urology, Helsingborg hospital, Helsingborg, Sweden
| | - Alexander Doyle
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Päivi Kannisto
- Department of Obstetrics and Gynecology, Department of Clinical Science, Skåne University Hospital, Lund University, Lund, Sweden
| | - Christian Brandt
- Department of Neurosurgery Lund, Department of Clinical Sciences Lund, Skåne University Hospital, Lund 22184, Sweden
| | - Emma Nilsson
- Division of Gastroenterology, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Lars B Dahlin
- Department of Translational Medicine-Hand Surgery, Lund University, Malmö, Sweden; Department of Hand Surgery, Skåne University Hospital, 20502 Malmö, Sweden; Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
| | - Johan Malm
- Department of Translational Medicine, Section for Clinical Chemistry, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden
| | - Adam Linder
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden
| | - Emma Niméus
- Division of Surgery, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden; Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Johan Malmström
- Division of infection medicine, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden; BioMS - National Infrastructure in Biological and Medical Mass Spectrometry, Department of Clinical Sciences Lund, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
6
|
Fan R, Liu H, Liang Q. Roles and Therapeutic Targeting of Exosomes in Sepsis-Induced Cardiomyopathy. J Cell Mol Med 2025; 29:e70559. [PMID: 40264381 PMCID: PMC12015131 DOI: 10.1111/jcmm.70559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Sepsis-induced cardiomyopathy (SICM) is a complex and fatal manifestation of sepsis, characterised by myocardial dysfunction that exacerbates the clinical prognosis in septic patients. While the pathophysiology of SICM remains incompletely understood, emerging evidence highlights the multifaceted functions of exosomes, small membrane-bound extracellular vesicles, in mediating the inflammatory responses and cardiac dysfunction involved in this condition. During sepsis, exosomes are secreted by various cells, such as cardiomyocytes, endothelial cells and macrophages, which serve as critical messengers, transferring proteins, lipids and RNA molecules that influence recipient cells, thus affecting cellular functions and disease progression. This review summarises the pathophysiology of SICM and the basics of exosomes and focuses on exosome-mediated mechanisms in SICM, including their role in inflammation, oxidative stress, mitochondrial dysfunction and myocardial injury, offering novel insights into the exosome-based therapeutic strategies in SICM.
Collapse
Affiliation(s)
- Rui Fan
- Graduate SchoolHeilongjiang University of Chinese MedicineHarbinChina
| | - Han Liu
- Graduate SchoolUniversity College LondonLondonUK
| | - Qun Liang
- Department of Critical Care MedicineFirst Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| |
Collapse
|
7
|
Dörtbudak MB, Demircioğlu M, Kapucuk FS. Micromeria congesta Alleviates LPS-Induced Inflammation, Apoptosis, Oxidative Stress and DNA Damage in Rat Heart and Kidneys. Vet Med Sci 2025; 11:e70264. [PMID: 40089897 PMCID: PMC11910719 DOI: 10.1002/vms3.70264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 03/17/2025] Open
Abstract
Life-threatening sepsis with high mortality and morbidity is an important cause of acute kidney injury and myocardial dysfunction. In this study, we investigated the protective effect of Micromeria congesta (MC) against kidney and heart damage caused by lipopolysaccharide (LPS) used as a sepsis model. Control, LPS, LPS + 25 mg/kg MC and LPS + 50 mg/kg MC groups were established from rats for the study. After the experiment, kidney and heart tissues obtained from the rats were stained with hematoxylin-eosin for histopathologic examination. Immunohistochemical staining was performed to determine inflammation, apoptosis, oxidative stress and DNA damage. IL-2 for inflammation, CASP-3 for apoptosis, HSP-27 for oxidative stress and 8-OHdG for DNA damage were used for immunopathologic examination. Histopathologic examination showed that the lesions in the kidney and heart tissues in the LPS group decreased with increasing doses of MC. Immunohistochemical examination showed that the expression of IL-2, CASP-3, HSP-27 and 8-OHdG was severe in the LPS group, but the severity of expression in these tissues decreased with increasing doses of MC. As a result of the study, it was histopathologically determined that MC reduced LPS-induced kidney and heart tissue damage. In addition, MC was found to protect against LPS by reducing LPS-induced inflammation, apoptosis, oxidative stress and DNA damage in kidney and heart tissue. In conclusion, it was seen that MC was effective in sepsis damage. However, it was concluded that MC could be an alternative in drug strategies developed for sepsis treatment with studies in vivo including more analyses.
Collapse
Affiliation(s)
| | - Muhammed Demircioğlu
- Department of Histology and Embryology, Institute of Health Sciences, Dicle University, Diyarbakir, Turkey
| | - Fuat Serkan Kapucuk
- Department of Pharmacology and Toxicology, Institute of Health Sciences, Harran University, Şanlıurfa, Turkey
| |
Collapse
|
8
|
Zakynthinos GE, Giamouzis G, Xanthopoulos A, Oikonomou E, Kalogeras K, Karavidas N, Dimeas IE, Gialamas I, Gounaridi MI, Siasos G, Vavuranakis M, Zakynthinos E, Tsolaki V. Septic Cardiomyopathy: Difficult Definition, Challenging Diagnosis, Unclear Treatment. J Clin Med 2025; 14:986. [PMID: 39941657 PMCID: PMC11818464 DOI: 10.3390/jcm14030986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Sepsis is a systemic inflammatory response syndrome of suspected or confirmed infectious origin, which frequently culminates in multiorgan failure, including cardiac involvement. Septic cardiomyopathy (SCM) remains a poorly defined clinical entity, lacking a formal or consensus definition and representing a significant knowledge gap in critical care medicine. It is an often-underdiagnosed complication of sepsis. The only widely accepted aspect of its definition is that SCM is a transient myocardial dysfunction occurring in patients with sepsis, which cannot be attributed to ischemia or pre-existing cardiac disease. The pathogenesis of SCM appears to be multifactorial, involving inflammatory cytokines, overproduction of nitric oxide, mitochondrial dysfunction, calcium homeostasis dysregulation, autonomic imbalance, and myocardial edema. Diagnosis primarily relies on echocardiography, with advanced tools such as tissue Doppler imaging (TDI) and global longitudinal strain (GLS) providing greater sensitivity for detecting subclinical dysfunction and guiding therapeutic decisions. Traditional echocardiographic findings, such as left ventricular ejection fraction measured by 2D echocardiography, often reflect systemic vasoplegia rather than intrinsic myocardial dysfunction, complicating accurate diagnosis. Right ventricular (RV) dysfunction, identified as a critical component of SCM in many studies, has multifactorial pathophysiology. Factors including septic cardiomyopathy itself, mechanical ventilation, hypoxemia, and hypercapnia-particularly in cases complicated by acute respiratory distress syndrome (ARDS)-increase RV afterload and exacerbate RV dysfunction. The prognostic value of cardiac biomarkers, such as troponins and natriuretic peptides, remains uncertain, as these markers primarily reflect illness severity rather than being specific to SCM. Treatment focuses on the early recognition of sepsis, hemodynamic optimization, and etiological interventions, as no targeted therapies currently exist. Emerging therapies, such as levosimendan and VA-ECMO, show potential in severe SCM cases, though further validation is needed. The lack of standardized diagnostic criteria, combined with the heterogeneity of sepsis presentations, poses significant challenges to the effective management of SCM. Future research should focus on developing cluster-based classification systems for septic shock patients by integrating biomarkers, echocardiographic findings, and clinical parameters. These advancements could clarify the underlying pathophysiology and enable tailored therapeutic strategies to improve outcomes for SCM patients.
Collapse
Affiliation(s)
- George E. Zakynthinos
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
| | - Grigorios Giamouzis
- Department of Cardiology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece; (G.G.); (A.X.)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 41110 Larissa, Greece; (G.G.); (A.X.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
| | - Nikitas Karavidas
- Critical Care Department, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Mezourlo, 41335 Larissa, Greece; (N.K.); (I.E.D.); (V.T.)
| | - Ilias E. Dimeas
- Critical Care Department, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Mezourlo, 41335 Larissa, Greece; (N.K.); (I.E.D.); (V.T.)
| | - Ioannis Gialamas
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
| | - Maria Ioanna Gounaridi
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
| | - Gerasimos Siasos
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, “Sotiria” Chest Diseases Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (G.E.Z.); (E.O.); (K.K.); (I.G.); (M.I.G.); (G.S.); (M.V.)
| | - Epaminondas Zakynthinos
- Critical Care Department, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Mezourlo, 41335 Larissa, Greece; (N.K.); (I.E.D.); (V.T.)
| | - Vasiliki Tsolaki
- Critical Care Department, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, Mezourlo, 41335 Larissa, Greece; (N.K.); (I.E.D.); (V.T.)
| |
Collapse
|
9
|
Dabrowski W, Pfortmueller CA, Kotfis K, Jaroszynski A, Gagos M, Plotek W, Malbrain MLNG. Is there a place for natural agents with anti-inflammatory and antioxidative properties in critically ill patients? Potential usefulness of Xanthohumol. Pharmacol Ther 2025; 266:108766. [PMID: 39637948 DOI: 10.1016/j.pharmthera.2024.108766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Multi-organ dysfunction is a major issue in critically ill patients, where a significant inflammatory response appears to be the primary factor driving the degree of organ impairment, which correlates with the extent of organ injury. The management of inflammation requires a multidisciplinary approach, including antibiotics for infection control, circulatory and respiratory support, and correction of coagulation abnormalities. However, the use of anti-inflammatory treatments is typically restricted to a selected group of medications, with their effectiveness remaining the subject of extensive debate. Xanthohumol (Xn), a natural compound extracted from hops, possesses strong anti-inflammatory and antioxidative properties, with a mild anti-coagulation effect. Its biological activity is related to the inhibition of different inflammatory pathways, reduction in cytokine production and secretion, and an increase in antioxidative enzyme activity. This review examined the potential use of Xn as an adjuvant in the treatment of various pathologies in critically ill patients.
Collapse
Affiliation(s)
- Wojciech Dabrowski
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Poland.
| | | | - Katarzyna Kotfis
- Department of Anaesthesiology, Intensive Care and Pain Management, Pomeranian Medical University of Szczecin, Poland
| | | | - Mariusz Gagos
- Department of Cell Biology, Maria Curie-Sklodowska University of Lublin, Poland
| | - Wlodzimierz Plotek
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Poland
| | - Manu L N G Malbrain
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Poland; Medical Data Management, Medaman, Geel, Belgium; International Fluid Academy, Lovenjoel, Belgium
| |
Collapse
|
10
|
Qiu Z, Zhou K, Qi Q, Chen W. Silencing fatty acid-binding protein 4 improved sepsis-induced myocardial dysfunction through anti-apoptotic and antioxidant effects by mammalian target of rapamycin signaling pathway. Cytojournal 2025; 22:8. [PMID: 39958886 PMCID: PMC11829329 DOI: 10.25259/cytojournal_157_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/23/2024] [Indexed: 02/18/2025] Open
Abstract
Objective One of the main complications of sepsis that is linked to poor clinical outcomes and high mortality is sepsis-induced myocardial dysfunction (SIMD). Fatty acid-binding protein 4 (FABP4) is a protein that is expressed in macrophages and adipose tissue and is involved in inflammation and apoptosis in various pathological processes. The purpose of this study was to investigate the role of FABP4 in SIMD. Material and Methods The H9c2 cell model of myocardial dysfunction induced by septicemia was established by lipopolysaccharide (LPS). Measurements of cell viability, apoptosis, reactive oxygen species levels, mitochondrial activity, and proinflammatory factor expression were used to assess FABP4's involvement in SIMD. In addition, the expression level of key proteins in the mammalian target of rapamycin (mTOR) signaling pathway was analyzed using Western blot. Finally, the combination of AZD-8055 further demonstrated the possibility of mTOR as a therapeutic target for SIMD. Results Silencing FABP4 expression drastically increased H9c2 cell viability and mitochondrial function. In addition, by upregulating B-cell lymphoma-2 (Bcl-2) and downregulating Bcl-2 associated X protein, FABP4 silencing improved LPS-induced anti-apoptosis of H9c2 cells. Finally, silencing FABP4 alleviated SIMD through the mTOR signaling pathway. However, the therapeutic effect was inhibited when FABP4 silencing was combined with the mTOR inhibitor AZD-8055. Conclusion Silencing FABP4 alleviates LPS-induced inflammatory response and apoptosis in H9c2 cells and enhances mitochondrial function through the mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhilei Qiu
- Department of Emergency, Hangzhou Xixi Hospital, Hangzhou, China
| | - Kexing Zhou
- Department of Emergency, Hangzhou Xixi Hospital, Hangzhou, China
| | - Qinyao Qi
- Department of Emergency, Hangzhou Xixi Hospital, Hangzhou, China
| | - Wei Chen
- Department of Emergency, Hangzhou Xixi Hospital, Hangzhou, China
| |
Collapse
|
11
|
Du X, Xiong F, Hou Y, Yu X, Pan P. Levosimendan for sepsis-induced myocardial dysfunction: friend or foe? Front Cardiovasc Med 2025; 11:1520596. [PMID: 39844909 PMCID: PMC11752121 DOI: 10.3389/fcvm.2024.1520596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) involves reversible myocardial dysfunction. The use of inotropes can restore adequate cardiac output and tissue perfusion, but conventional inotropes, such as dobutamine and adrenaline, have limited efficacy in such situations. Levosimendan is a novel inotrope that acts in a catecholamine-independent manner. However, study results regarding the treatment of SIMD with levosimendan are inconsistent, and the use of levosimendan is highly controversial. In this review, we summarized the therapeutic mechanisms of levosimendan in SIMD and considered recent research on how to improve the efficacy of levosimendan in SIMD. We also analyzed the potential and limitations of levosimendan for the treatment of SIMD to provide ideas for future clinical trials and the clinical application of levosimendan in SIMD.
Collapse
Affiliation(s)
- Xinxin Du
- Cardiac Intensive Care Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Fang Xiong
- Department of Critical Care Medicine, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Yafei Hou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyou Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Pengfei Pan
- Department of Critical Care Medicine, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| |
Collapse
|
12
|
Wiger CW, Ranheim T, Arnesen H, Vaage J, Pischke SE, Yndestad A, Stensløkken K, Torp M. TLR4 Inhibition Attenuated LPS-Induced Proinflammatory Signaling and Cytokine Release in Mouse Hearts and Cardiomyocytes. Immun Inflamm Dis 2025; 13:e70133. [PMID: 39853914 PMCID: PMC11760985 DOI: 10.1002/iid3.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Sepsis is associated with myocardial injury and early mortality. The innate immune receptor Toll-like receptor 4 (TLR4) can recognize pathogen-associated-molecular-patterns (PAMPs) and damage-associated molecular patterns (DAMPs); the latter are released during tissue injury. We hypothesized that TLR4 inhibition reduces proinflammatory signaling and cytokine release in: (1) LPS or Escherichia coli-treated isolated mouse heart; (2) LPS-treated mouse primary adult cardiomyocytes; and (3) the isolated heart during ischemia-reperfusion. METHODS Isolated C57BL/6N male mouse hearts were perfused for 120 min, with either LPS, E. coli, with and without CLI-095 (TLR4 inhibitor). Primary adult mouse cardiomyocytes were treated with LPS or LPS + CLI-095. Isolated hearts, exposed to 35 min of global ischemia, were treated with either vehicle or CLI-095 during reperfusion. Infarct size was quantified by triphenyltetrazolium staining. Cytokine expression was analyzed with ELISA, western blot analysis, and qPCR. RESULTS In isolated hearts, E. coli increased the expression of proinflammatory cytokines (IL-6 and CXCL2), which was not attenuated with TLR4 inhibition. TLR4 inhibition reduced expression (p = 0.004) and release of IL-6 (p < 0.0001) in LPS-exposed isolated hearts. LPS activated the nuclear-factor κ-light-chain-enhancer of activated B cells signaling pathway (NF-κB) in primary adult cardiomyocytes. Moreover, TLR4 inhibition reduced LPS-induced mRNA expression and release of IL-6 in primary adult cardiomyocytes. Isolated hearts treated with CLI-095 during reperfusion after ischemia (induced DAMPs release) showed reduced infarct size (39 ± 17% to 26 ± 8%, p = 0.034) and decreased IL-6 release (p = 0.006). CONCLUSION Inhibition of TLR4 reduced proinflammatory signaling and cytokine release in LPS-treated and ischemia-reperfused isolated mouse hearts and in primary adult murine cardiomyocytes.
Collapse
Affiliation(s)
- Christine W. Wiger
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Henriette Arnesen
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jarle Vaage
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
- Department of Research and Innovation, Division of Emergencies and Critical CareOslo University HospitalOsloNorway
| | | | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Kåre‐Olav Stensløkken
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - May‐Kristin Torp
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Østfold Hospital TrustGrålumNorway
| |
Collapse
|
13
|
Ghazaly HF, Aly AAA, Tammam AS, Hassan MM, Hammad SS, Mahmoud NM, Hemaida TS. Influence of liberal versus conservative oxygen therapies on the hemodynamic parameters of mechanically ventilated patients with sepsis: a randomized clinical trial. BMC Anesthesiol 2024; 24:469. [PMID: 39707209 PMCID: PMC11660434 DOI: 10.1186/s12871-024-02838-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND There is no significant evidence verifying the efficacy of liberal versus conservative oxygen therapy on hemodynamics in patients with sepsis. We investigated how liberal and conservative oxygen therapy influenced stroke volume, cardiac output, and vasopressor needs in patients with sepsis undergoing mechanical ventilation. METHODS This randomized clinical trial included 106 patients with an admission diagnosis of infection, a Sequential Organ Failure Assessment (SOFA) score of two points or higher and required invasive mechanical ventilation for at least 72 h. Patients were randomly assigned to one of two oxygenation strategies: liberal (n = 53) with a target SpO2 of ≥ 96% or conservative (n = 53) with a target SpO2 of 88-92%. Transthoracic Doppler echocardiography was done twice to measure stroke volume and cardiac output, initially upon enrollment in the trial and then 72 h later. The primary outcome was stroke volume. Secondary outcomes were cardiac output, vasopressor use, mechanical ventilation duration, ICU stay length, and adverse events. RESULTS Stroke volume and cardiac output measurements did not differ significantly between research groups after 72 h of oxygenation treatment (p = 0.459 and 0.637, respectively). Forty-five patients (84.9%) in the conservative oxygen therapy group needed vasopressors to maintain their mean arterial pressure above 65 mmHg, whereas 35 patients (66.0%) in the liberal group did (p = 0.024). A multivariate logistic regression analysis of the independent variables for vasopressor requirements revealed that patients in the conservative oxygen group were 3.83 times more likely to require vasopressors (AOR = 3.83, 95% CI: 1.31-11.18, p = 0.014) than those in the liberal group. Older patients (AOR = 1.03, 95% CI: 1.01-1.07, p = 0.038) and those with higher SOFA scores (AOR = 1.36, CI: 1.09-1.68, P = 0.005) were significantly more likely to need vasopressors. CONCLUSIONS Liberal or conservative oxygen therapy did not influence stroke volume or cardiac output measurements in mechanically ventilated patients with sepsis. Patients in the conservative oxygen group were more likely to require vasopressors than those in the liberal group. TRIAL REGISTRATION This study was approved by the Ethics Committee of Aswan University Hospital (approval number: Aswu/460/5/20) (registration date: 05/05/2020) and registered on ClinicalTrials.gov (NCT04824703) (03/30/2021).
Collapse
Affiliation(s)
- Huda F Ghazaly
- Anesthesia and Surgical Intensive Care Department, Faculty of Medicine, Aswan University, Aswan, Egypt.
| | - Ahmed Alsaied A Aly
- Anesthesia and Surgical Intensive Care Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Ahmed S Tammam
- Anesthesia and Surgical Intensive Care Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Mahmoud M Hassan
- Anesthesia and Surgical Intensive Care Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Soudy S Hammad
- Anesthesia and Surgical Intensive Care Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Naggeh M Mahmoud
- Cardiology Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Tarek S Hemaida
- Anesthesia and Surgical Intensive Care Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| |
Collapse
|
14
|
Tian T, Yu Q, Yang D, Zhang X, Zhang C, Li J, Luo T, Zhang K, Lv X, Wang Y, Wang H, Li H. Endothelial α 1-adrenergic receptor activation improves cardiac function in septic mice via PKC-ERK/p38MAPK signaling pathway. Int Immunopharmacol 2024; 141:112937. [PMID: 39182270 DOI: 10.1016/j.intimp.2024.112937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Cardiomyopathy is particularly common in septic patients. Our previous studies have shown that activation of the alpha 1 adrenergic receptor (α1-AR) on cardiomyocytes inhibits sepsis-induced myocardial dysfunction. However, the role of cardiac endothelial α1-AR in septic cardiomyopathy has not been determined. Here, we identified α1-AR expression in mouse and human endothelial cells and showed that activation of α1-AR with phenylephrine (PE) improved cardiac function and survival by preventing cardiac endothelial injury in septic mice. Mechanistically, activating α1-AR with PE decreased the expression of ICAM-1, VCAM-1, iNOS, E-selectin, and p-p38MAPK, while promoting PKC and ERK1/2 phosphorylation in LPS-treated endothelial cells. These effects were abolished by a PKC inhibitor or α1-AR antagonist. PE also reduced p65 nuclear translocation, but this suppression is not blocked by PKC inhibition. Treatment with U0126 (a specific ERK1/2 inhibitor) reversed the effects of PE on p38MAPK phosphorylation. Our results demonstrate that cardiac endothelial α1-AR activation prevents sepsis-induced myocardial dysfunction in mice by inhibiting the endothelial injury via PKC-ERK/p38MAPK signaling pathway and a PKC-independent inhibition of p65 nuclear translocation. These findings offer a new perspective for septic patients with cardiac dysfunction by inhibiting cardiac endothelial cell injury through α1-AR activation.
Collapse
Affiliation(s)
- Tian Tian
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Qing Yu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Duomeng Yang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xue Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chanjuan Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jianling Li
- Department of Anesthesiology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China
| | - Keke Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiuxiu Lv
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Huadong Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hongmei Li
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
15
|
Wu J, Ai T, He P, Shi Q, Li Y, Zhang Z, Chen M, Huang Z, Wu S, Chen W, Han J. Cecal necroptosis triggers lethal cardiac dysfunction in TNF-induced severe SIRS. Cell Rep 2024; 43:114778. [PMID: 39325617 DOI: 10.1016/j.celrep.2024.114778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Tumor necrosis factor (TNF) induces systemic inflammatory response syndrome (SIRS), and severe SIRS can serve as a model for studying animal death caused by organ failure. Through strategic cecectomy, we demonstrate that necroptosis in the cecum initiates the death process in TNF-treated mice, but it is not the direct cause of death. Instead, we show that it is the cardiac dysfunction downstream of cecum damage that ultimately leads to the death of TNF-treated mice. By in vivo and ex vivo physiological analyses, we reveal that TNF and the damage-associated molecular patterns (DAMPs) released from necroptotic cecal cells jointly target cardiac endothelial cells, triggering caspase-8 activation and subsequent cardiac endothelial damage. Cardiac endothelial damage is a primary cause of the deterioration of diastolic function in the heart of TNF-treated mice. Our research provides insights into the pathophysiological process of TNF-induced lethality.
Collapse
Affiliation(s)
- Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Ai
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Peng He
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Qilin Shi
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangxin Li
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ziguan Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanze Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518000, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
16
|
Killu K, Patino-Sutton C, Kysh L, Castriotta R, Oropello J, Huerta L, Engracia D, Merchant K, Wee CP, Cortessis VK. The association between integrating echocardiography use in the management of septic shock patients and outcomes in the intensive care unit: a systematic review and meta-analysis. J Ultrasound 2024:10.1007/s40477-024-00958-w. [PMID: 39419883 DOI: 10.1007/s40477-024-00958-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/18/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVES Septic shock in critically ill patients can increases morbidity and mortality. We aimed to study the effect on outcomes when integrating point of care (POC) echocardiography in the management of septic shock patients in the Intensive Care Unit (ICU) who are being treated according to the Surviving Sepsis Campaign (SSC) guidelines. METHODS An electronic search of MEDLINE through PubMed, clinical trials.gov and google scholar was conducted for the period from January 1990-January 2024 to identify studies of septic shock adult and pediatric patients in the ICU managed according to SSC guidelines with or without POC echocardiography. Three reviewers extracted data independent of each other. Cochrane collaboration tool was used for bias assessment. Random effect meta-analysis used to pool data. RESULTS A total of 1701 articles identified. Seven studies included in the final report with a total of 3885 patients. POC echocardiography guided septic shock management was associated with lower in-hospital and 28-day mortality (sOR = 0.82 [95%CI: 0.71-0.95], p = 0.01), more frequent initiation of inotropic support (sOR = 2.42 [95%CI 1.92-3.03], p < 0.0001) and shorter time to achieve lactate clearance (SMD = - 0.87 h [95%CI - 1.23 h to - 0.51 h], p < 0.0001). Summary estimates did not achieve significance for effect of POC echocardiography on 24-h fluid intake (SMD = - 2.11 ml [95%CI - 5.93 ml to 1.72 ml], p = 0.28) on mechanical ventilation-free days (SMD = 0.03 days [95%CI - 0.04 to 0.10], p = 0.94). Shock reversal time analysis was less meaningful due to the small number of studies reporting outcome. CONCLUSIONS POC echocardiography guided management in septic shock patients in the ICU can lead to a decrease in mortality, increase in initiation of inotropic support, and a decrease in lactate clearance time. Larger cohort studies and data collection and analysis are needed for further understanding and optimizing standardization of protocols for POC echocardiography use in septic shock patients in the ICU.
Collapse
Affiliation(s)
- Keith Killu
- Keck School of Medicine, Pulmonary Critical Care and Sleep Division, Department of Internal Medicine, University of Southern California, 2020 Zonal Ave., IRD #720, Los Angeles, CA, 90033, USA.
| | - Cecilia Patino-Sutton
- Keck School of Medicine, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Lynn Kysh
- Keck School of Medicine, Norris Medical Library, University of Southern California, Los Angeles, CA, USA
- Davis Library, Univercity of California, Davis, CA, 95616, USA
| | - Richard Castriotta
- Keck School of Medicine, Pulmonary Critical Care and Sleep Division, Department of Internal Medicine, University of Southern California, 2020 Zonal Ave., IRD #720, Los Angeles, CA, 90033, USA
| | - John Oropello
- The Ichan School of Medicine at the Mount Sinai Hospital, New York, NY, USA
| | - Luis Huerta
- Keck School of Medicine, Pulmonary Critical Care and Sleep Division, Department of Internal Medicine, University of Southern California, 2020 Zonal Ave., IRD #720, Los Angeles, CA, 90033, USA
| | - Dominic Engracia
- Keck School of Medicine, Pulmonary Critical Care and Sleep Division, Department of Internal Medicine, University of Southern California, 2020 Zonal Ave., IRD #720, Los Angeles, CA, 90033, USA
| | - Karim Merchant
- Keck School of Medicine, Pulmonary Critical Care and Sleep Division, Department of Internal Medicine, University of Southern California, 2020 Zonal Ave., IRD #720, Los Angeles, CA, 90033, USA
| | - Choo Phei Wee
- Keck School of Medicine, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Victoria Kristence Cortessis
- Keck School of Medicine, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Wang Z, Li L, Yang S, Li Z, Zhang P, Shi R, Zhou X, Tang X, Li Q. Possible mechanisms of SARS-CoV-2-associated myocardial fibrosis: reflections in the post-pandemic era. Front Microbiol 2024; 15:1470953. [PMID: 39444690 PMCID: PMC11497467 DOI: 10.3389/fmicb.2024.1470953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Since December 2019, coronavirus disease 2019 (COVID-19) has been spreading worldwide with devastating immediate or long-term effects on people's health. Although the lungs are the primary organ affected by COVID-19, individuals infected with SARS-CoV-2 also develop systemic lesions involving multiple organs throughout the body, such as the cardiovascular system. Emerging evidence reveals that COVID-19 could generate myocardial fibrosis, termed "COVID-19-associated myocardial fibrosis." It can result from the activation of fibroblasts via the renin-angiotensin-aldosterone system (RAAS), transforming growth factor-β1 (TGF-β1), microRNAs, and other pathways, and can also occur in other cellular interactions with SARS-CoV-2, such as immunocytes, endothelial cells. Nonetheless, to gain a more profound insight into the natural progression of COVID-19-related myocardial fibrosis, additional investigations are necessary. This review delves into the underlying mechanisms contributing to COVID-19-associated myocardial fibrosis while also examining the antifibrotic potential of current COVID-19 treatments, thereby offering guidance for future clinical trials of these medications. Ultimately, we propose future research directions for COVID-19-associated myocardial fibrosis in the post-COVID-19 era, such as artificial intelligence (AI) telemedicine. We also recommend that relevant tests be added to the follow-up of COVID-19 patients to detect myocardial fibrosis promptly.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luwei Li
- Department of Pediatric Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Third Clinical Medical College of Zhengzhou University, Zhengzhou, China
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojuan Tang
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Maiorov I, Bagrov K, Efraim R, Ankri Eliyahu G, Livneh A, Landesberg A. MMP-8 causes leftward shift in end-diastolic pressure-volume relationship and may explain the development of diastolic dysfunction in septic cardiomyopathy. Am J Physiol Heart Circ Physiol 2024; 327:H1098-H1111. [PMID: 39178029 DOI: 10.1152/ajpheart.00240.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Septic cardiomyopathy (SCM) with diastolic dysfunction carries a poor prognosis, and the mechanisms underlying the development of diastolic dysfunction remain unclear. Matrix metalloproteinase-8 (MMP-8) is released from neutrophils and degrades collagen I. MMP-8 levels correlate with SCM severity. We scrutinized, for the first time, the direct impact of MMP-8 on cardiac systolic and diastolic functions. Isolated rat hearts were perfused with Krebs-Henseleit solution in a Langendorff setup with computer-controlled filling pressures of both ventricles in an isovolumetric regime. The end-diastolic pressure (EDP) varied periodically between 3 and 20 mmHg. After baseline recordings, MMP-8 (100 µg/mL) was added to the perfusion. Short-axis views of both ventricles were continuously acquired by echocardiography. MMP-8 perfusion resulted in a progressive decline in peak systolic pressures (Psys) in both ventricles, but without significant changes in their end-systolic pressure-area relationships (ESPARs). Counterintuitively, conspicuous leftward shifts of the end-diastolic pressure-area relationships (EDPARs) were observed in both ventricles. The left ventricle (LV) end-diastolic area (EDA) decreased by 32.8 ± 5.7% (P = 0.008) at an EDP of 10.5 ± 0.4 mmHg, when LV Psys dropped by 20%. The decline of Psys was primarily due to the decrease in EDA, and restoring the baseline EDA by increasing EDP recovered 81.33 ± 5.87% of the pressure drop. Collagen I generates tensile (eccentric) stress, and its degradation by MMP-8 causes end-diastolic pressure-volume relationship (EDPVR) leftward shift, resulting in diastolic and systolic dysfunctions. The diastolic dysfunction explains the clinically observed fluid unresponsiveness, whereas the decrease in end-diastolic volume (EDV) diminishes the systolic functions. MMP-8 can explain the development of SCM with diastolic dysfunction.NEW & NOTEWORTHY MMP-8, released from activated neutrophils and macrophages, is markedly elevated in sepsis, correlating with sepsis severity and mortality. MMP-8 targets collagen I of the cardiac ECM and induces diastolic dysfunction with fluid unresponsiveness, associated with decreased EDV, reduced sarcomere length, and diminished systolic function. Unlike other MMPs that predominantly cleave collagen-III and contribute to cardiac dilatation, thereby increasing sarcomere length, MMP-8 leads to a leftward shift in the EDPVR, resulting in diastolic and systolic dysfunctions.
Collapse
Affiliation(s)
- Ida Maiorov
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Konstantin Bagrov
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Roy Efraim
- Cardiology Department, Rambam Health Care Campus, Haifa, Israel
| | - Galit Ankri Eliyahu
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Amit Livneh
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| | - Amir Landesberg
- Cardiovascular Research, Faculty of Biomedical Engineering, Technion-IIT, Haifa, Israel
| |
Collapse
|
19
|
Liang X, Hu X, Li J, Zhang B, Gu T, Wang H, Zhang M, Xia X, Guan S, Shangguan W, Miao S, Wang W, Zhang H, Zhao Z, Wang L. m6A methylation in myocardial tissue of septic mice analyzed using MeRIP/m6A-sequencing and RNA-sequencing. Funct Integr Genomics 2024; 24:173. [PMID: 39320434 DOI: 10.1007/s10142-024-01452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Septic cardiomyopathy is a secondary myocardial injury caused by sepsis. N6-methyl-adenosine (m6A) modification is involved in the pathological progression of septic cardiomyopathy; however, the pathological mechanism remains unclear. In this study, we identified the overall m6A modification pattern in septic myocardial injury and determined its potential interactions with differentially expressed genes (DEGs). A sepsis mouse model exhibiting septic symptoms and myocardial tissue damage was induced by lipopolysaccharide (LPS). LPS-induced septic myocardial tissues and control myocardial tissues were subjected to methylated RNA immunoprecipitation sequencing and RNA sequencing to screen for differentially expressed m6A peaks and DEGs. We identified 859 significantly m6A-modified genes in septic myocardial tissues, including 432 upregulated and 427 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to explore the biological importance of differentially expressed m6A methylated genes and DEGs. Differentially expressed m6A methylated genes were enriched in immune- and inflammation-related pathways. Conjoint analysis revealed co-expression of differentially expressed m6A genes and DEGs, including genes that were upregulated or downregulated and those showing opposite trends. High expression of m6A-related genes (WTAP and IGF2BP2), interleukin-17, and interleukin-17 pathway-related genes (MAPK11 and TRAF3IP2) was verified using reverse transcription-quantitative PCR. We confirmed the presence of m6A modification of the transcriptome and m6A-mediated gene expression in septic myocardial tissues.
Collapse
Affiliation(s)
- Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xiaotong Hu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jiao Li
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, 300121, China
| | - Boyang Zhang
- Department of Emergency Medicine, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Tianshu Gu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hualing Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Mingzhong Zhang
- Department of Traditional Chinese Medicine, People's Hospital of Linqing, Linqing Shandong, 252600, China
| | - Xiaodong Xia
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Siyu Guan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wenfeng Shangguan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shuai Miao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Weiding Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Hao Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| | - Lijun Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
20
|
Orso D, Federici N, Lio C, Mearelli F, Bove T. Hemodynamic goals in sepsis and septic shock resuscitation: An umbrella review of systematic reviews and meta-analyses with trial sequential analysis. Aust Crit Care 2024; 37:818-826. [PMID: 38609748 DOI: 10.1016/j.aucc.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/03/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
OBJECTIVE The objective of this study was to verify whether any parameter among those used as the target for haemodynamic optimisation (e.g., mean arterial pressure, central venous oxygen saturation, systolic or diastolic dysfunction, CO2 gap, lactates, right ventricular dysfunction, and PvaCO2/CavO2 ratio) is correlated with mortality in an undifferentiated population with sepsis or septic shock. METHODS An umbrella review, searching MEDLINE, the Cochrane Database of Systematic Reviews, Health Technology Assessment Database, and the JBI Database of Systematic Reviews and Implementation Reports, was performed. We included systematic reviews and meta-analyses enrolling a population of unselected patients with sepsis or septic shock. The main outcome was mortality. Two authors conducted data extraction and risk-of-bias assessments independently. We used a random-effects model to pool binary and continuous data and summarised estimates of effect using equivalent odds ratios (eORs). We used the ROBIS tool to assess risk of bias and the assessment of multiple systematic reviews 2 score to assess global quality. DATA SYNTHESIS 17 systematic reviews and meta-analyses (15 828 patients) were included in the quantitative analysis. Diastolic dysfunction (eOR: 1.42; 95% confidence interval [CI]: 1.14-1.76), PvaCO2/CavO2 ratio (eOR: 2.15; 95% CI: 1.37-3.37), and CO2 gap (eOR: 1.86; 95% CI: 1.07-3.25) showed a significant correlation with mortality. Lactates were the parameter with highest inconsistency (I2 = 92%). Central venous oxygen saturation and right ventricle dysfunction showed significant statistical excess test of significance (p-value = 0.009 and 0.005, respectively). None of the considered parameters showed statistically significant publication bias. CONCLUSIONS According to this umbrella review, diastolic dysfunction is the haemodynamic variable that is most closely linked to the prognosis of septic patients. The PvaCO2/CavO2 ratio and the CO2gap are significantly related to the mortality of septic patients, but the poor quality of evidence or the low number of cases, studied so far, limit their clinical applicability. CLINICAL TRIAL REGISTRATION PROSPERO: International prospective register of systematic reviews, 2023, CRD42023432813 (Available from: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023432813).
Collapse
Affiliation(s)
- Daniele Orso
- Department of Anesthesia and Intensive Care Medicine, ASUFC University Hospital of Udine, Udine, Italy.
| | - Nicola Federici
- Department of Anesthesia and Intensive Care Medicine, ASUFC University Hospital of Udine, Udine, Italy
| | - Cristina Lio
- Department of Anesthesia and Intensive Care Medicine, ASUFC University Hospital of Udine, Udine, Italy; Department of Medical Sciences (DAME), University of Udine, Udine, Italy
| | - Filippo Mearelli
- Department of Internal Medicine, ASUGI University Hospital of Trieste, Trieste, Italy
| | - Tiziana Bove
- Department of Anesthesia and Intensive Care Medicine, ASUFC University Hospital of Udine, Udine, Italy; Department of Medical Sciences (DAME), University of Udine, Udine, Italy
| |
Collapse
|
21
|
Srdić T, Đurašević S, Lakić I, Ružičić A, Vujović P, Jevđović T, Dakić T, Đorđević J, Tosti T, Glumac S, Todorović Z, Jasnić N. From Molecular Mechanisms to Clinical Therapy: Understanding Sepsis-Induced Multiple Organ Dysfunction. Int J Mol Sci 2024; 25:7770. [PMID: 39063011 PMCID: PMC11277140 DOI: 10.3390/ijms25147770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Sepsis-induced multiple organ dysfunction arises from the highly complex pathophysiology encompassing the interplay of inflammation, oxidative stress, endothelial dysfunction, mitochondrial damage, cellular energy failure, and dysbiosis. Over the past decades, numerous studies have been dedicated to elucidating the underlying molecular mechanisms of sepsis in order to develop effective treatments. Current research underscores liver and cardiac dysfunction, along with acute lung and kidney injuries, as predominant causes of mortality in sepsis patients. This understanding of sepsis-induced organ failure unveils potential therapeutic targets for sepsis treatment. Various novel therapeutics, including melatonin, metformin, palmitoylethanolamide (PEA), certain herbal extracts, and gut microbiota modulators, have demonstrated efficacy in different sepsis models. In recent years, the research focus has shifted from anti-inflammatory and antioxidative agents to exploring the modulation of energy metabolism and gut microbiota in sepsis. These approaches have shown a significant impact in preventing multiple organ damage and mortality in various animal sepsis models but require further clinical investigation. The accumulation of this knowledge enriches our understanding of sepsis and is anticipated to facilitate the development of effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Tijana Srdić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Siniša Đurašević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Iva Lakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Aleksandra Ružičić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Predrag Vujović
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tanja Jevđović
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tamara Dakić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Jelena Đorđević
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| | - Tomislav Tosti
- Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sofija Glumac
- School of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Zoran Todorović
- School of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (S.G.); (Z.T.)
| | - Nebojša Jasnić
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (T.S.); (S.Đ.); (I.L.); (A.R.); (P.V.); (T.J.); (T.D.); (J.Đ.)
| |
Collapse
|
22
|
Yu HP, Liu FC, Chung YK, Alalaiwe A, Sung CT, Fang JY. Nucleic acid-based nanotherapeutics for treating sepsis and associated organ injuries. Theranostics 2024; 14:4411-4437. [PMID: 39113804 PMCID: PMC11303080 DOI: 10.7150/thno.98487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
In recent years, gene therapy has been made possible with the success of nucleic acid drugs against sepsis and its related organ dysfunction. Therapeutics based on nucleic acids such as small interfering RNAs (siRNAs), microRNAs (miRNAs), messenger RNAs (mRNAs), and plasmid DNAs (pDNAs) guarantee to treat previously undruggable diseases. The advantage of nucleic acid-based therapy against sepsis lies in the development of nanocarriers, achieving targeted and controlled gene delivery for improved efficacy with minimal adverse effects. Entrapment into nanocarriers also ameliorates the poor cellular uptake of naked nucleic acids. In this study, we discuss the current state of the art in nanoparticles for nucleic acid delivery to treat hyperinflammation and apoptosis associated with sepsis. The optimized design of the nanoparticles through physicochemical property modification and ligand conjugation can target specific organs-such as lung, heart, kidney, and liver-to mitigate multiple sepsis-associated organ injuries. This review highlights the nanomaterials designed for fabricating the anti-sepsis nanosystems, their physicochemical characterization, the mechanisms of nucleic acid-based therapy in working against sepsis, and the potential for promoting the therapeutic efficiency of the nucleic acids. The current investigations associated with nanoparticulate nucleic acid application in sepsis management are summarized in this paper. Noteworthily, the potential application of nanotherapeutic nucleic acids allows for a novel strategy to treat sepsis. Further clinical studies are required to confirm the findings in cell- and animal-based experiments. The capability of large-scale production and reproducibility of nanoparticle products are also critical for commercialization. It is expected that numerous anti-sepsis possibilities will be investigated for nucleic acid-based nanotherapeutics in the future.
Collapse
Affiliation(s)
- Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Yu-Kuo Chung
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Calvin T. Sung
- Department of Dermatology, University of California, Irvine, United States
| | - Jia-You Fang
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
| |
Collapse
|
23
|
Cater DT, Meyers BA, Mitra S, Bhattacharya S, Machado RF, Serrano R, Rowan CM, Gaston B, Vlachos P. NOVEL ECHOCARDIOGRAM ANALYSIS OF CARDIAC DYSFUNCTION IS ASSOCIATED WITH MORTALITY IN PEDIATRIC SEPSIS. Shock 2024; 62:26-31. [PMID: 38661156 PMCID: PMC11833790 DOI: 10.1097/shk.0000000000002359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
ABSTRACT Objectives: The objective of our study was to semiautomatically generate echocardiogram indices in pediatric sepsis using novel algorithms and determine which indices were associated with mortality. We hypothesized that strain and diastolic indices would be most associated with mortality. Design: Retrospective cohort study of children with sepsis from 2017 to 2022. Survivors and nonsurvivors were compared for echocardiogram indices. Multivariate Cox proportional hazard models were constructed for our primary outcome of in-hospital mortality. Linear regression was performed for secondary outcomes, which included multiple composite 28-day outcomes. Results: Of the 54 patients in the study, 9 (17%) died. Multiple echocardiogram indices of both right (RV) and left ventricles (LV) were associated with in-hospital mortality [RV GLS adjusted hazard ratio (aHR): 1.16 (1.03-1.29), P = 0.011; RV global longitudinal early diastolic strain rate (GLSre) aHR: 0.24 (0.07 to 0.75), P = 0.014; LV GLSre aHR: 0.33 (0.11-0.97), P = 0.044]. Impairment in GLS was associated with fewer ventilator-free days [RV GLS β-coefficient: -0.47 (-0.84 to -0.10), P = 0.013; LV GLS β-coefficient -0.62 (-1.07 to -0.17), P = 0.008], organ-support free days [RV GLS β-coefficient: -0.49 (-0.87 to -0.11), P = 0.013; LV GLS β-coefficient: -0.64 (-1.10 to -0.17), P = 0.008], and days free from ICU [RV GLS β-coefficient: -0.42 (-0.79 to -0.05), P = 0.026; LV GLS β-coefficient: -0.58 (-1.03 to -0.13), P = 0.012]. Systolic indices were not associated with mortality in this cohort. Conclusion: Our study demonstrates the feasibility of obtaining echocardiogram indices in a semiautomatic method using our algorithms. We showed that abnormal strain is associated with worse outcomes in a cohort of children with sepsis.
Collapse
Affiliation(s)
- Daniel T Cater
- Division of Critical Care, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis Indiana
| | - Brett A Meyers
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana
| | - Shailee Mitra
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana
| | | | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ryan Serrano
- Division of Pediatric Cardiology, Loma Linda University Children's Hospital and Medical Center, Loma Linda, California
| | - Courtney M Rowan
- Division of Critical Care, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis Indiana
| | - Benjamin Gaston
- Riley Hospital for Children and the Herman B. Wells Center for Pediatric Research, Indianapolis, Indiana
| | - Pavlos Vlachos
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana
| |
Collapse
|
24
|
Casper E, El Wakeel L, Sabri N, Khorshid R, Fahmy SF. Melatonin: A potential protective multifaceted force for sepsis-induced cardiomyopathy. Life Sci 2024; 346:122611. [PMID: 38580195 DOI: 10.1016/j.lfs.2024.122611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Sepsis is a life-threatening condition manifested by organ dysfunction caused by a dysregulated host response to infection. Lung, brain, liver, kidney, and heart are among the affected organs. Sepsis-induced cardiomyopathy is a common cause of death among septic patients. Sepsis-induced cardiomyopathy is characterized by an acute and reversible significant decline in biventricular both systolic and diastolic function. This is accompanied by left ventricular dilatation. The pathogenesis underlying sepsis-induced cardiomyopathy is multifactorial. Hence, targeting an individual pathway may not be effective in halting the extensive dysregulated immune response. Despite major advances in sepsis management strategies, no effective pharmacological strategies have been shown to treat or even reverse sepsis-induced cardiomyopathy. Melatonin, namely, N-acetyl-5-methoxytryptamine, is synthesized in the pineal gland of mammals and can also be produced in many cells and tissues. Melatonin has cardioprotective, neuroprotective, and anti-tumor activity. Several literature reviews have explored the role of melatonin in preventing sepsis-induced organ failure. Melatonin was found to act on different pathways that are involved in the pathogenesis of sepsis-induced cardiomyopathy. Through its antimicrobial, anti-inflammatory, and antioxidant activity, it offers a potential role in sepsis-induced cardiomyopathy. Its antioxidant activity is through free radical scavenging against reactive oxygen and nitrogen species and modulating the expression and activity of antioxidant enzymes. Melatonin anti-inflammatory activities control the overactive immune system and mitigate cytokine storm. Also, it mitigates mitochondrial dysfunction, a major mechanism involved in sepsis-induced cardiomyopathy, and thus controls apoptosis. Therefore, this review discusses melatonin as a promising drug for the management of sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Eman Casper
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Lamia El Wakeel
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Nagwa Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Ramy Khorshid
- Department of Cardiovascular and Thoracic Surgery, Ain Shams University Hospital, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Sarah F Fahmy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
25
|
Duignan SM, Lakshminrusimha S, Armstrong K, de Boode WP, El-Khuffash A, Franklin O, Molloy EJ. Neonatal sepsis and cardiovascular dysfunction I: mechanisms and pathophysiology. Pediatr Res 2024; 95:1207-1216. [PMID: 38044334 DOI: 10.1038/s41390-023-02926-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 12/05/2023]
Abstract
The highest incidence of sepsis across all age groups occurs in neonates leading to substantial mortality and morbidity. Cardiovascular dysfunction frequently complicates neonatal sepsis including biventricular systolic and/or diastolic dysfunction, vasoregulatory failure, and pulmonary arterial hypertension. The haemodynamic response in neonatal sepsis can be hyperdynamic or hypodynamic and the underlying pathophysiological mechanisms are heterogeneous. The diagnosis and definition of both neonatal sepsis and cardiovascular dysfunction complicating neonatal sepsis are challenging and not consensus-based. Future developments in neonatal sepsis management will be facilitated by common definitions and datasets especially in neonatal cardiovascular optimisation. IMPACT: Cardiovascular dysfunction is common in neonatal sepsis but there is no consensus-based definition, making calculating the incidence and designing clinical trials challenging. Neonatal cardiovascular dysfunction is related to the inflammatory response, which can directly target myocyte function and systemic haemodynamics.
Collapse
Affiliation(s)
- Sophie M Duignan
- Department of Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | | | - Kathryn Armstrong
- Children's Heart Centre, BC Children's Hospital, Vancouver, BC, Canada
| | - Willem P de Boode
- Department of Neonatology, Radboud University Medical Center, Radboud Institute for Health Sciences, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Afif El-Khuffash
- School of Medicine, Department of Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Orla Franklin
- Department of Paediatric Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
- Discipline of Paediatrics, Trinity College, The University of Dublin, Trinity Research in Childhood (TRiCC) & Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College, The University of Dublin, Trinity Research in Childhood (TRiCC) & Trinity Translational Medicine Institute (TTMI), Dublin, Ireland.
- Department of Neonatology, Children's Health Ireland at Crumlin, Dublin, Ireland.
- Department of Neonatology, Coombe Women and Infants University Hospital, Dublin, Ireland.
- Paediatric Neurodisability, Children's Health Ireland at Tallaght, Dublin, Ireland.
| |
Collapse
|
26
|
Sargsyan Z, Srivastava SD, Triant VA, Ghoshhajra BB. Case 9-2024: An 84-Year-Old Man with a Fall. N Engl J Med 2024; 390:1129-1139. [PMID: 38507756 DOI: 10.1056/nejmcpc2312731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Affiliation(s)
- Zaven Sargsyan
- From the Department of Medicine, Baylor College of Medicine, Houston (Z.S.); and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Massachusetts General Hospital, and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Harvard Medical School - both in Boston
| | - Sunita D Srivastava
- From the Department of Medicine, Baylor College of Medicine, Houston (Z.S.); and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Massachusetts General Hospital, and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Harvard Medical School - both in Boston
| | - Virginia A Triant
- From the Department of Medicine, Baylor College of Medicine, Houston (Z.S.); and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Massachusetts General Hospital, and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Harvard Medical School - both in Boston
| | - Brian B Ghoshhajra
- From the Department of Medicine, Baylor College of Medicine, Houston (Z.S.); and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Massachusetts General Hospital, and the Departments of Surgery (S.D.S.), Medicine (V.A.T.), and Radiology (B.B.G.), Harvard Medical School - both in Boston
| |
Collapse
|
27
|
Chen B, Li YF, Fang Z, Cai WY, Tian ZQ, Li D, Wang ZM. Epigallocatechin-3-gallate protects sepsis-induced myocardial dysfunction by inhibiting the nuclear factor-κB signaling pathway. Heliyon 2024; 10:e27163. [PMID: 38449632 PMCID: PMC10915574 DOI: 10.1016/j.heliyon.2024.e27163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) has become one of the most lethal complications of sepsis, while the treatment was limited by a shortage of pertinent drugs. Epigallocatechin-3-gallate (EGCG) is the highest content of active substances in green tea, and its application in cardiovascular diseases has broad prospects. This study was conducted to test the hypothesis that EGCG was able to inhibit lipopolysaccharide (LPS) induced myocardial dysfunction and investigate the underlying molecular mechanisms. The cardiac systolic function was assessed by echocardiography. The cardiomyocyte apoptosis was determined by TUNEL staining. The expression of inflammatory factors and apoptosis-related protein, cardiac markers were examined by Western Blot and qRT-PCR. EGCG effectively improve LPS-induced cardiac function damage, enhance left ventricular systolic function, and restore myocardial cell vitality. It can effectively inhibit the upregulation of TLR4 expression induced by LPS and inhibit IκB α/NF- κB/p65 signaling pathway, thereby inhibiting cardiomyocyte apoptosis and improving myocarditis. In conclusion, EGCG protects against SIMD through anti-inflammatory and anti-apoptosis effects; it was mediated by the inhibition of the TLR4/NF-κB signal pathway. Our results demonstrated that EGCG might be a possible medicine for SIMD prevention and treatment.
Collapse
Affiliation(s)
- Bei Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Ya-Fei Li
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, Jiangsu Province, China
| | - Zhang Fang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Wen-Yi Cai
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Zhi-Qiang Tian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Dianfu Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Ze-Mu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| |
Collapse
|
28
|
Kim JH, Lee JH. Effect of miR-412-5p-loaded exosomes in H9c2 cardiomyocytes via the MAPK pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:755-760. [PMID: 38645496 PMCID: PMC11024402 DOI: 10.22038/ijbms.2024.75590.16365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/15/2024] [Indexed: 04/23/2024]
Abstract
Objectives MicroRNAs (miRNAs) are small non-coding RNAs that function in all biological processes. Recent findings suggest that exosomes, which are small vesicles abundantly secreted by various cell types, can transport miRNAs to target cells. Here, we elucidated the effect of miRNA-loaded exosomes on lipopolysaccharide (LPS)-induced inflammation in H9c2 cardiomyocytes. Materials and Methods Exosomes were isolated from mesenchymal stem cells (MSC) and loaded with miR-412-5p. Additionally, the effect of the miR-412-5p-loaded exosomes on LPS-induced inflammation in H9c2 cardiomyocytes was evaluated by assessing the levels of nitric oxide (NO), reactive oxygen species (ROS), and prostaglandin E2 (PGE2). The expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), inflammatory cytokines, and mitogen-activated protein kinase (MAPK) signaling factors was evaluated using reverse transcription-quantitative PCR and western blotting. Results miR-412-5p-loaded exosomes inhibited LPS-induced secretion of inflammatory mediators (NO, PGE2, and ROS), pro-inflammatory cytokines (IL-1β and IL-6), and COX-2 and iNOS expression. Additionally, miR-412-5p-loaded exosomes significantly decreased the expression of MAPK signaling molecules, including p-extracellular signal-regulated kinase (ERK), p-p38, and p-Jun kinase (JNK), in H9c2 cardiomyocytes. Conclusion These findings showed that miR-412-5p-loaded exosomes ameliorated LPS-induced inflammation in H9c2 cardiomyocytes by inhibiting COX-2 and iNOS expression, inflammatory mediators, and pro-inflammatory cytokines via the MAPK pathway. The findings indicate that miR-412-5p-loaded exosomes may be effective for the prevention of myocardial injury.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Anesthesiology and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - June Hwan Lee
- Department of Energy Information Technology, Fareast University, 76-32, Daehak-gil, Gamgok-myeon, Eumseong-gun, Chungcheongbuk-do 27601, Republic of Korea
| |
Collapse
|
29
|
Lukić I, Mihić D, Varžić SC, Relatić KS, Zibar L, Loinjak D, Ćurić ŽB, Klobučar L, Maričić L. Septic Cardiomyopathy. Rev Cardiovasc Med 2024; 25:23. [PMID: 39077653 PMCID: PMC11262393 DOI: 10.31083/j.rcm2501023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 07/31/2024] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis-induced myocardial dysfunction represents reversible myocardial dysfunction which ultimately results in left ventricular dilatation or both, with consequent loss of contractility. Studies on septic cardiomyopathy report a wide range of prevalence ranging from 10% to 70%. Myocardial damage occurs as a result of weakened myocardial circulation, direct myocardial depression, and mitochondrial dysfunction. Mitochondrial dysfunction is the leading problem in the development of septic cardiomyopathy and includes oxidative phosphorylation, production of reactive oxygen radicals, reprogramming of energy metabolism, and mitophagy. Echocardiography provides several possibilities for the diagnosis of septic cardiomyopathy. Systolic and diastolic dysfunction of left ventricular is present in 50-60% of patients with sepsis. Right ventricular dysfunction is present in 50-55% of cases, while isolated right ventricular dysfunction is present in 47% of cases. Left ventricle (LV) diastolic dysfunction is very common in septic shock, and it represents an early biomarker, it has prognostic significance. Right ventricular dysfunction associated with sepsis patients with worse early prognosis. Global longitudinal stress and magnetic resonance imaging (MRI) of the heart are sufficiently sensitive methods, but at the same time MRI of the heart is difficult to access in intensive care units, especially when dealing with critically ill patients. Previous research has identified two biomarkers as a result of the integrated mitochondrial response to stress, and these are fibroblast growth factor-21 (FGF-21) and growth differentiation factor-15 (GDF-15). Both of the mentioned biomarkers can be easily quantified in serum or plasma, but they are difficult to be specific in patients with multiple comorbidities. Mitochondrial dysfunction is also associated with reduced levels of miRNA (microRNA), some research showed significance of miRNA in sepsis-induced myocardial dysfunction, but further research is needed to determine the clinical significance of these molecules in septic cardiomyopathy. Therapeutic options in the treatment of septic cardiomyopathy are not specific, and include the optimization of hemodynamic parameters and the use of antibiotic thera-pies with targeted action. Future research aims to find mechanisms of targeted action on the initial mechanisms of the development of septic cardiomyopathy.
Collapse
Affiliation(s)
- Ivana Lukić
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Heart and Vascular Diseases, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Damir Mihić
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Pulmology and Intensive Care Medicine, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Silvija Canecki Varžić
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Endocrinology, University Hospital Centre Hospital Osijek, 31000 Osijek, Croatia
| | - Kristina Selthofer Relatić
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Heart and Vascular Diseases, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Lada Zibar
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Nephrology, University Hospital Merkur, Zagreb, 10000 Zagreb, Croatia
| | - Domagoj Loinjak
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Pulmology and Intensive Care Medicine, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Željka Breškić Ćurić
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Internal Medicine, General Hospital Vinkovci, 32100 Vinkovci, Croatia
| | - Lucija Klobučar
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Heart and Vascular Diseases, University Hospital Centre Osijek, 31000 Osijek, Croatia
| | - Lana Maričić
- Faculty of Medicine, University J. J. Strossmayer in Osijek, 31000 Osijek, Croatia
- Department of Heart and Vascular Diseases, University Hospital Centre Osijek, 31000 Osijek, Croatia
| |
Collapse
|
30
|
Marcos-Vidal JM, González R, Merino M, Higuera E, García C. Sedation for Patients with Sepsis: Towards a Personalised Approach. J Pers Med 2023; 13:1641. [PMID: 38138868 PMCID: PMC10744994 DOI: 10.3390/jpm13121641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
This article looks at the challenges of sedoanalgesia for sepsis patients, and argues for a personalised approach. Sedation is a necessary part of treatment for patients in intensive care to reduce stress and anxiety and improve long-term prognoses. Sepsis patients present particular difficulties as they are at increased risk of a wide range of complications, such as multiple organ failure, neurological dysfunction, septic shock, ARDS, abdominal compartment syndrome, vasoplegic syndrome, and myocardial dysfunction. The development of any one of these complications can cause the patient's rapid deterioration, and each has distinct implications in terms of appropriate and safe forms of sedation. In this way, the present article reviews the sedative and analgesic drugs commonly used in the ICU and, placing special emphasis on their strategic administration in sepsis patients, develops a set of proposals for sedoanalgesia aimed at improving outcomes for this group of patients. These proposals represent a move away from simplistic approaches like avoiding benzodiazepines to more "objective-guided sedation" that accounts for a patient's principal pathology, as well as any comorbidities, and takes full advantage of the therapeutic arsenal currently available to achieve personalised, patient-centred treatment goals.
Collapse
Affiliation(s)
- José Miguel Marcos-Vidal
- Department of Anesthesiology and Critical Care, Universitary Hospital of Leon, 24071 Leon, Spain; (R.G.); (M.M.); (E.H.); (C.G.)
| | | | | | | | | |
Collapse
|
31
|
Fan Y, Guan B, Xu J, Zhang H, Yi L, Yang Z. Role of toll-like receptor-mediated pyroptosis in sepsis-induced cardiomyopathy. Biomed Pharmacother 2023; 167:115493. [PMID: 37734261 DOI: 10.1016/j.biopha.2023.115493] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Sepsis, a life-threatening dysregulated status of the host response to infection, can cause multiorgan dysfunction and mortality. Sepsis places a heavy burden on the cardiovascular system due to the pathological imbalance of hyperinflammation and immune suppression. Myocardial injury and cardiac dysfunction caused by the aberrant host responses to pathogens can lead to cardiomyopathy, one of the most critical complications of sepsis. However, many questions about the specific mechanisms and characteristics of this complication remain to be answered. The causes of sepsis-induced cardiac dysfunction include abnormal cardiac perfusion, myocardial inhibitory substances, autonomic dysfunction, mitochondrial dysfunction, and calcium homeostasis dysregulation. The fight between the host and pathogens acts as the trigger for sepsis-induced cardiomyopathy. Pyroptosis, a form of programmed cell death, plays a critical role in the progress of sepsis. Toll-like receptors (TLRs) act as pattern recognition receptors and participate in innate immune pathways that recognize damage-associated molecular patterns as well as pathogen-associated molecular patterns to mediate pyroptosis. Notably, pyroptosis is tightly associated with cardiac dysfunction in sepsis and septic shock. In line with these observations, induction of TLR-mediated pyroptosis may be a promising therapeutic approach to treat sepsis-induced cardiomyopathy. This review focuses on the potential roles of TLR-mediated pyroptosis in sepsis-induced cardiomyopathy, to shed light on this promising therapeutic approach, thus helping to prevent and control septic shock caused by cardiovascular disorders and improve the prognosis of sepsis patients.
Collapse
Affiliation(s)
- Yixuan Fan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Jianxing Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Liang Yi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhixu Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
32
|
Guarracino F, Cortegiani A, Antonelli M, Behr A, Biancofiore G, Del Gaudio A, Forfori F, Galdieri N, Grasselli G, Paternoster G, Rocco M, Romagnoli S, Sardo S, Treskatsch S, Tripodi VF, Tritapepe L. The role of beta-blocker drugs in critically ill patients: a SIAARTI expert consensus statement. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2023; 3:41. [PMID: 37872608 PMCID: PMC10591347 DOI: 10.1186/s44158-023-00126-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND The role of β-blockers in the critically ill has been studied, and data on the protective effects of these drugs on critically ill patients have been repeatedly reported in the literature over the last two decades. However, consensus and guidelines by scientific societies on the use of β-blockers in critically ill patients are still lacking. The purpose of this document is to support the clinical decision-making process regarding the use of β-blockers in critically ill patients. The recipients of this document are physicians, nurses, healthcare personnel, and other professionals involved in the patient's care process. METHODS The Italian Society of Anesthesia, Analgesia, Resuscitation and Intensive Care (SIAARTI) selected a panel of experts and asked them to define key aspects underlying the use of β-blockers in critically ill adult patients. The methodology followed by the experts during this process was in line with principles of modified Delphi and RAND-UCLA methods. The experts developed statements and supportive rationales in the form of informative text. The overall list of statements was subjected to blind votes for consensus. RESULTS The literature search suggests that adrenergic stress and increased heart rate in critically ill patients are associated with organ dysfunction and increased mortality. Heart rate control thus seems to be critical in the management of the critically ill patient, requiring careful clinical evaluation aimed at both the differential diagnosis to treat secondary tachycardia and the treatment of rhythm disturbance. In addition, the use of β-blockers for the treatment of persistent tachycardia may be considered in patients with septic shock once hypovolemia has been ruled out. Intravenous application should be the preferred route of administration. CONCLUSION β-blockers protective effects in critically ill patients have been repeatedly reported in the literature. Their use in the acute treatment of increased heart rate requires understanding of the pathophysiology and careful differential diagnosis, as all causes of tachycardia should be ruled out and addressed first.
Collapse
Affiliation(s)
- Fabio Guarracino
- Cardiothoracic and Vascular Anesthesia and Intensive Care, Anesthesia and Resuscitation Department, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Andrea Cortegiani
- Department of Surgical, Oncological and Oral Science (Di.Chir.On.S.), University of Palermo, Palermo, Italy.
- Department of Anesthesia, Intensive Care and Emergency, Policlinico Paolo Giaccone, University of Palermo, 90127, Palermo, Italy.
| | - Massimo Antonelli
- Department of Emergency, Anesthesiological and Resuscitation Sciences, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Astrid Behr
- Operative Unit of Anesthesia and Resuscitation, Hospital of Camposampiero, Padua, Italy
| | - Giandomenico Biancofiore
- Anesthesia and Resuscitation Transplants, Department of Medical Pathology Surgical, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - Alfredo Del Gaudio
- Emergency Department, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesco Forfori
- Anesthesia and Intensive Care, Anesthesia and Resuscitation Department, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Nicola Galdieri
- General Cardiac Surgery Unit, Critical Area Department, Ospedale Dei Colli, Naples, Italy
| | - Giacomo Grasselli
- Department of Anesthesia, Resuscitation and Emergency, IRCCS Ca' Granda Foundation, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Medical-Surgical and Transplant Pathophysiology, University of Milan, Milan, Italy
| | | | - Monica Rocco
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefano Romagnoli
- Anesthesia and Intensive Care Section, Department of Health Sciences, University of Florence, Florence, Italy
- Department of Anesthesia and Intensive Care, Careggi University Hospital, Florence, Italy
| | - Salvatore Sardo
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Sascha Treskatsch
- Department of Anesthesiology and Intensive Care Medicine, Charité Universitätsmedizin Berlin, Freie Universität and Humboldt-Universität Zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Vincenzo Francesco Tripodi
- Anesthesia and Intensive Care Unit, Department of Surgery, University Hospital "Gaetano Martino", Messina, Italy
| | - Luigi Tritapepe
- Anesthesia and Resuscitation Unit, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| |
Collapse
|
33
|
Zhang L, Qi D, Peng M, Meng B, Wang X, Zhang X, Zuo Z, Li L, Wang Z, Zou W, Hu Z, Qian Z. Decoding molecular signature on heart of septic mice with distinct left ventricular ejection fraction. iScience 2023; 26:107825. [PMID: 37736036 PMCID: PMC10509301 DOI: 10.1016/j.isci.2023.107825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023] Open
Abstract
Dysregulated cardiac function after sepsis in intensive care unit is known to predict poor long-term outcome and increase mortality. Their pathological feature and molecular mechanism remain unclear. We observed that septic patients with depressed left ventricular ejection fraction (LVEF) have the highest in-hospital and 28 days mortality comparing to patients with hyperdynamic LVEF or with heart failure with preserved LVEF. Echocardiograms reveal that survivors post cecum ligation and puncture (CLP) on rodents have stable LVEF and non-survivors have fluctuated LVEF at CLP early phase. CLP-induced mice fall into three groups based on LVEF 24 h post-surgery: high-, low-, and normal-LVEF. Transcriptomic and proteomic analyses identify jointly and distinctively changed genes, proteins and biologically essential pathways in left ventricles from three CLP groups. Notably, transmission electron microscopy shows different mitochondrial and sarcomere defects associated with LVEF variances. Together, this study systematically characterizes the molecular, morphological, and functional alterations in CLP-induced cardiac injury.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Desheng Qi
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Milin Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Binbin Meng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinrun Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaolei Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhihong Zuo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Zhanwen Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| | - Wenxuan Zou
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhonghua Hu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital) Changsha, Changsha 410008, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Changsha 410008, China
| |
Collapse
|
34
|
Giugni FR, Aiello VD, Faria CS, Pour SZ, Cunha MDP, Giugni MV, Pinesi HT, Ledesma FL, Morais CE, Ho YL, Sztajnbok J, de Morais Fernezlian S, Ferraz da Silva LF, Mauad T, Ferreira Alves VA, Hilário do Nascimento Saldiva P, Antonangelo L, Dolhnikoff M, Duarte-Neto AN. Understanding yellow fever-associated myocardial injury: an autopsy study. EBioMedicine 2023; 96:104810. [PMID: 37757571 PMCID: PMC10550587 DOI: 10.1016/j.ebiom.2023.104810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Yellow fever (YF) is a viral hemorrhagic fever, endemic in parts of South America and Africa. There is scarce evidence about the pathogenesis of the myocardial injury. The objective of this study is to evaluate the cardiac pathology in fatal cases of YF. METHODS This retrospective autopsy study included cases from the São Paulo (Brazil) epidemic of 2017-2019. We reviewed medical records and performed cardiac tissue histopathological evaluation, electron microscopy, immunohistochemical assays, RT-qPCR for YF virus (YFV)-RNA, and proteomics analysis on inflammatory and endothelial biomarkers. FINDINGS Seventy-three confirmed YF cases with a median age of 48 (34-60) years were included. We observed myocardial fibrosis in 68 (93.2%) patients; cardiomyocyte hypertrophy in 68 (93.2%); endothelial alterations in 67 (91.8%); fiber necrosis in 50 (68.5%); viral myocarditis in 9 (12.3%); and secondary myocarditis in 5 (6.8%). Four out of five patients with 17DD vaccine-associated viscerotropic disease presented with myocarditis. The cardiac conduction system showed edema, hemorrhages and endothelial fibrinoid necrosis. Immunohistochemistry detected CD68-positive inflammatory interstitial cells and YFV antigens in endothelial and inflammatory cells. YFV-RNA was detected positive in 95.7% of the cardiac samples. The proteomics analysis demonstrated that YF patients had higher levels of multiple inflammatory and endothelial biomarkers in comparison to cardiovascular controls, and higher levels of interferon gamma-induced protein 10 (IP-10) in comparison to sepsis (p = 0.01) and cardiovascular controls (p < 0.001) in Dunn test. INTERPRETATION Myocardial injury is frequent in severe YF, due to multifactorial mechanisms, including direct YFV-mediated damage, endothelial cell injury, and inflammatory response, with a possible prominent role for IP-10. FUNDING This study was funded by Fundação de Amparo à Pesquisa do Estado de São Paulo, Bill and Melinda Gates Foundation, Conselho Nacional de Desenvolvimento Científico e Tecnológico, Coordenação de Aperfeiçoamento de Pessoal de Nível Superior.
Collapse
Affiliation(s)
- Fernando Rabioglio Giugni
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Instituto do Coração InCor, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vera Demarchi Aiello
- Instituto do Coração InCor, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Caroline Silverio Faria
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Shahab Zaki Pour
- Laboratório de Evolução Molecular e Bioinformática, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil
| | - Marielton Dos Passos Cunha
- Laboratório de Evolução Molecular e Bioinformática, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil
| | - Melina Valdo Giugni
- Instituto do Coração InCor, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Henrique Trombini Pinesi
- Instituto do Coração InCor, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Felipe Lourenço Ledesma
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Carolina Esteves Morais
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Yeh-Li Ho
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Serviço de Verificação de Óbitos da Capital (SVOC), Universidade de São Paulo, São Paulo, SP, Brazil
| | - Thais Mauad
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Leila Antonangelo
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Amaro Nunes Duarte-Neto
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
35
|
Jakobsson G, Papareddy P, Andersson H, Mulholland M, Bhongir R, Ljungcrantz I, Engelbertsen D, Björkbacka H, Nilsson J, Manea A, Herwald H, Ruiz-Meana M, Rodríguez-Sinovas A, Chew M, Schiopu A. Therapeutic S100A8/A9 blockade inhibits myocardial and systemic inflammation and mitigates sepsis-induced myocardial dysfunction. Crit Care 2023; 27:374. [PMID: 37773186 PMCID: PMC10540409 DOI: 10.1186/s13054-023-04652-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND AND AIMS The triggering factors of sepsis-induced myocardial dysfunction (SIMD) are poorly understood and are not addressed by current treatments. S100A8/A9 is a pro-inflammatory alarmin abundantly secreted by activated neutrophils during infection and inflammation. We investigated the efficacy of S100A8/A9 blockade as a potential new treatment in SIMD. METHODS The relationship between plasma S100A8/A9 and cardiac dysfunction was assessed in a cohort of 62 patients with severe sepsis admitted to the intensive care unit of Linköping University Hospital, Sweden. We used S100A8/A9 blockade with the small-molecule inhibitor ABR-238901 and S100A9-/- mice for therapeutic and mechanistic studies on endotoxemia-induced cardiac dysfunction in mice. RESULTS In sepsis patients, elevated plasma S100A8/A9 was associated with left-ventricular (LV) systolic dysfunction and increased SOFA score. In wild-type mice, 5 mg/kg of bacterial lipopolysaccharide (LPS) induced rapid plasma S100A8/A9 increase and acute LV dysfunction. Two ABR-238901 doses (30 mg/kg) administered intraperitoneally with a 6 h interval, starting directly after LPS or at a later time-point when LV dysfunction is fully established, efficiently prevented and reversed the phenotype, respectively. In contrast, dexamethasone did not improve cardiac function compared to PBS-treated endotoxemic controls. S100A8/A9 inhibition potently reduced systemic levels of inflammatory mediators, prevented upregulation of inflammatory genes and restored mitochondrial function in the myocardium. The S100A9-/- mice were protected against LPS-induced LV dysfunction to an extent comparable with pharmacologic S100A8/A9 blockade. The ABR-238901 treatment did not induce an additional improvement of LV function in the S100A9-/- mice, confirming target specificity. CONCLUSION Elevated S100A8/A9 is associated with the development of LV dysfunction in severe sepsis patients and in a mouse model of endotoxemia. Pharmacological blockade of S100A8/A9 with ABR-238901 has potent anti-inflammatory effects, mitigates myocardial dysfunction and might represent a novel therapeutic strategy for patients with severe sepsis.
Collapse
Affiliation(s)
- Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Lund, Sweden
- Cardiac Inflammation Research Group, Clinical Research Center, 91:12, Jan Waldenströms Gata 35, 21 428, Malmö, Sweden
| | | | - Henrik Andersson
- Department of Anaesthesia and Intensive Care, Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Megan Mulholland
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Ravi Bhongir
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | | | - Harry Björkbacka
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Adrian Manea
- Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Heiko Herwald
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Rodríguez-Sinovas
- Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Michelle Chew
- Department of Anaesthesia and Intensive Care, Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Alexandru Schiopu
- Department of Translational Medicine, Lund University, Lund, Sweden.
- Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania.
- Department of Internal Medicine, Skane University Hospital, Lund, Sweden.
- Cardiac Inflammation Research Group, Clinical Research Center, 91:12, Jan Waldenströms Gata 35, 21 428, Malmö, Sweden.
| |
Collapse
|
36
|
Verra C, Mohammad S, Alves GF, Porchietto E, Coldewey SM, Collino M, Thiemermann C. Baricitinib protects mice from sepsis-induced cardiac dysfunction and multiple-organ failure. Front Immunol 2023; 14:1223014. [PMID: 37781388 PMCID: PMC10536262 DOI: 10.3389/fimmu.2023.1223014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/07/2023] [Indexed: 10/03/2023] Open
Abstract
Sepsis is one of the major complications of surgery resulting in high morbidity and mortality, but there are no specific therapies for sepsis-induced organ dysfunction. Data obtained under Gene Expression Omnibus accession GSE131761 were re-analyzed and showed an increased gene expression of Janus Kinase 2 (JAK2) and Signal Transducer and Activator of Transcription 3 (STAT3) in the whole blood of post-operative septic patients. Based on these results, we hypothesized that JAK/STAT activation may contribute to the pathophysiology of septic shock and, hence, investigated the effects of baricitinib (JAK1/JAK2 inhibitor) on sepsis-induced cardiac dysfunction and multiple-organ failure (MOF). In a mouse model of post-trauma sepsis induced by midline laparotomy and cecal ligation and puncture (CLP), 10-week-old male (n=32) and female (n=32) C57BL/6 mice received baricitinib (1mg/kg; i.p.) or vehicle at 1h or 3h post-surgery. Cardiac function was assessed at 24h post-CLP by echocardiography in vivo, and the degree of MOF was analyzed by determination of biomarkers in the serum. The potential mechanism underlying both the cardiac dysfunction and the effect of baricitinib was analyzed by western blot analysis in the heart. Trauma and subsequent sepsis significantly depressed the cardiac function and induced multiple-organ failure, associated with an increase in the activation of JAK2/STAT3, NLRP3 inflammasome and NF- κβ pathways in the heart of both male and female animals. These pathways were inhibited by the administration of baricitinib post the onset of sepsis. Moreover, treatment with baricitinib at 1h or 3h post-CLP protected mice from sepsis-induced cardiac injury and multiple-organ failure. Thus, baricitinib may be repurposed for trauma-associated sepsis.
Collapse
Affiliation(s)
- Chiara Verra
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Shireen Mohammad
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Elisa Porchietto
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Sina Maren Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
37
|
Karolczak K, Watala C. Estradiol as the Trigger of Sirtuin-1-Dependent Cell Signaling with a Potential Utility in Anti-Aging Therapies. Int J Mol Sci 2023; 24:13753. [PMID: 37762053 PMCID: PMC10530977 DOI: 10.3390/ijms241813753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Aging entails the inevitable loss of the structural and functional integrity of cells and tissues during the lifetime. It is a highly hormone-dependent process; although, the exact mechanism of hormone involvement, including sex hormones, is unclear. The marked suppression of estradiol synthesis during menopause suggests that the hormone may be crucial in maintaining cell lifespan and viability in women. Recent studies also indicate that the same may be true for men. Similar anti-aging features are attributed to sirtuin 1 (SIRT1), which may possibly be linked at the molecular level with estradiol. This finding may be valuable for understanding the aging process, its regulation, and possible prevention against unhealthy aging. The following article summarizes the initial studies published in this field with a focus on age-associated diseases, like cancer, cardiovascular disease and atherogenic metabolic shift, osteoarthritis, osteoporosis, and muscle damage, as well as neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Kamil Karolczak
- Department of Haemostatic Disorders, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland;
| | | |
Collapse
|
38
|
Zheng P, Wang X, Guo T, Gao W, Huang Q, Yang J, Gao H, Liu Q. Cardiac troponin as a prognosticator of mortality in patients with sepsis: A systematic review and meta-analysis. Immun Inflamm Dis 2023; 11:e1014. [PMID: 37773717 PMCID: PMC10515504 DOI: 10.1002/iid3.1014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND The impact of cardiac troponin on the short-term and long-term prognosis of patients with sepsis remains uncertain. Therefore, we conducted a meta-analysis to investigate the role of cardiac troponin as a potential indicator for sepsis mortality. METHODS We performed a comprehensive search for articles published before November 2022 using Google Scholar, PubMed, and Web of Science. Inclusion criteria for the studies were: (1) investigation of cardiac troponin, and (2) investigation of sepsis. Exclusion criteria included: (1) inability to obtain or calculate hazard ratio (HR) and 95% confidence interval (CI) for the relationship between cardiac troponin level and sepsis mortality, and (2) reviews, meta-analyses, and case reports. Analysis of HRs and 95% CIs for the association between cardiac troponin level and sepsis mortality was conducted using STATA 12.0 software. RESULTS Our study included 24 prospective studies (comprising 20,457 sepsis patients) and 4 retrospective studies (comprising 1416 sepsis patients). Meta-analysis demonstrated that elevated cardiac troponin levels were significantly associated with increased sepsis mortality using a random effects model (HR = 1.57, 95% CI 1.41-1.75). Moreover, elevated cardiac troponin levels were also significantly associated with increased hospital mortality of sepsis (HR = 1.35, 95% CI 1.19-1.53) and long-term mortality of sepsis (HR = 1.96, 95% CI 1.51-2.55) using the random effects model. CONCLUSIONS Overall, our finding revealed that elevated cardiac troponin for sepsis patients was a predictor of hospital and long-term mortality. Clinicians may treat septic patients with elevated cardiac troponin more cautious to avoid extra death. Moreover, large clinical studies are warranted to validate this association.
Collapse
Affiliation(s)
- Peiqiu Zheng
- Department of EmergencyLiyang Hospital of Chinese MedicineChangzhouJiangsuChina
| | - Xing Wang
- Department of Critical Care MedicineAffiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Tao Guo
- Department of EmergencyAffiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Wei Gao
- Department of Critical Care MedicineJiangsu Province Hospital on Integration of Chinese and Western MedicineNanjingJiangsuChina
| | - Qiang Huang
- Department of EmergencyLiyang Hospital of Chinese MedicineChangzhouJiangsuChina
| | - Jie Yang
- Department of Critical Care MedicineLiyang Hospital of Chinese MedicinChangzhouJiangsuChina
| | - Hui Gao
- Department of Critical Care MedicineLiyang Hospital of Chinese MedicinChangzhouJiangsuChina
| | - Qian Liu
- Department of Critical Care MedicineLiyang Hospital of Chinese MedicinChangzhouJiangsuChina
| |
Collapse
|
39
|
Hadi SMH, Majeed S, Ghafil FA, Altoraihi K, Hadi NR. Xanthohumol ameliorates cardiac injury induced by sepsis in a mice model: role of toll-like receptor 4. J Med Life 2023; 16:1105-1110. [PMID: 37900069 PMCID: PMC10600665 DOI: 10.25122/jml-2023-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/10/2023] [Indexed: 10/31/2023] Open
Abstract
Sepsis, a life-threatening condition arising from infection, often results in multi-organ failure, including cardiac dysfunction. This study investigated Xanthohumol, a natural compound, and its potential mechanism of action to enhance heart function following sepsis. A total of twenty-four adult male Swiss albino mice were allocated randomly to one of four equal groups (n=6): sham, CLP, vehicle Xanthohumol the same amount of DMSO injected IP 10 minutes before the CLP, and Xanthohumol group (0.4 mg/kg of Xanthohumol administered IP before the CLP process). Toll-like receptor 4, pro-inflammatory mediators, anti-inflammatory markers, oxidative stress indicators, apoptosis markers, and serum cardiac damage biomarkers were measured in the cardiac tissue using ELISA. Data with normal distribution were analyzed using t-test and ANOVA tests (p<0.05). In comparison to the sham group, the sepsis group had significantly higher levels of TLR-4, IL-6, TNF-α, MIF, F2-isoprostane, caspase-3, cTn-I, and CK-MB, while the pre-treated group with Xanthohumol had significantly lower levels (p<0.05) of these markers than the sepsis group. Bcl-2 showed no significant difference in Xanthohumol pre-treated group relative to the sepsis group, while IL-10 was significantly elevated. Xanthohumol dramatically reduced cardiac tissue injury (p<0.05) relative to the CLP group. By blocking the downstream signal transduction pathways of TLR-4 and NF-kB, Xanthohumol was shown to lessen cardiac damage in male mice during CLP-induced polymicrobial sepsis.
Collapse
Affiliation(s)
| | - Sahar Majeed
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Fadhaa Abdulameer Ghafil
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Kaswer Altoraihi
- Medical College, Department of Pharmacology and Therapeutics, University of Kufa, Najaf, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| |
Collapse
|
40
|
Hadi SMH, Majeed S, Ghafil FA, Altoraihi K, Hadi NR. Effect of Sulforaphane on cardiac injury induced by sepsis in a mouse model: Role of toll-like receptor 4. J Med Life 2023; 16:1120-1126. [PMID: 37900081 PMCID: PMC10600659 DOI: 10.25122/jml-2023-0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/21/2023] [Indexed: 10/31/2023] Open
Abstract
As sepsis is associated with a 50% increase in mortality, sepsis-induced cardiomyopathy has become a critical topic. A multidisciplinary approach is required for the diagnosis and treatment of septic cardiomyopathy. This study looked at Sulforaphane, a natural product that aims to evaluate cardiac function after sepsis, and its likely mechanism of action. Twenty-four adult male Swiss albino mice were randomly divided into 4 equal groups (n=6): sham, CLP, vehicle Sulforaphane (the same amount of DMSO injected IP one hour before the CLP), and Sulforaphane group (one hour before the CLP, a 5mg/kg dose of Sulforaphane was injected). Cardiac tissue levels of toll-like receptor 4 (TLR-4), pro-inflammatory mediators, anti-inflammatory markers, oxidative stress markers, apoptosis markers, and serum cardiac damage biomarkers were assessed using ELISA. Statistical analyses, including t-tests and ANOVA tests, were performed with a significance level of 0.05 for normally distributed data. Compared to the sham group, the sepsis group had significantly elevated levels of TLR-4, IL-6, TNF-α, MIF, F2-isoprostane, caspase-3, cTn-I, and CK-MB (p<0.05). In contrast, the Sulforaphane pre-treated group demonstrated significantly lower levels of these markers (p<0.05). Additionally, Bcl-2 levels were significantly reduced (p<0.05) in the Sulforaphane group. Sulforaphane administration also significantly attenuated cardiac tissue injury (p<0.05). The findings suggest that Sulforaphane can decrease heart damage in male mice during CLP-induced polymicrobial sepsis by suppressing TLR-4/NF-kB downstream signal transduction pathways.
Collapse
Affiliation(s)
| | - Sahar Majeed
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Fadhaa Abdulameer Ghafil
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| | - Kaswer Altoraihi
- Medical College, Department of Pharmacology and Therapeutics, University of Kufa, Najaf, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Najaf, Iraq
| |
Collapse
|
41
|
Thirunavukkarasu M, Swaminathan S, Kemerley A, Pradeep SR, Lim ST, Accorsi D, Wilson R, Campbell J, Saad I, Yee SP, Palesty JA, McFadden DW, Maulik N. Role of Pellino-1 in Inflammation and Cardioprotection following Severe Sepsis: A Novel Mechanism in a Murine Severe Sepsis Model †. Cells 2023; 12:1527. [PMID: 37296648 PMCID: PMC10252528 DOI: 10.3390/cells12111527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVES Intra-abdominal sepsis is commonly diagnosed in the surgical population and remains the second most common cause of sepsis overall. Sepsis-related mortality remains a significant burden in the intensive care unit despite advances in critical care. Nearly a quarter of the deaths in people with heart failure are caused by sepsis. We have observed that overexpression of mammalian Pellino-1 (Peli1), an E3 ubiquitin ligase, causes inhibition of apoptosis, oxidative stress, and preservation of cardiac function in a myocardial infarction model. Given these manifold applications, we investigated the role of Peli1 in sepsis using transgenic and knockout mouse models specific to this protein. Therefore, we aimed to explore further the myocardial dysfunction seen in sepsis through its relation to the Peli 1 protein by using the loss of function and gain-of-function strategy. METHODS A series of genetic animals were created to understand the role of Peli1 in sepsis and the preservation of heart function. Wild-type, global Peli1 knock out (Peli1-/-), cardiomyocyte-specific Peli1 deletion (CP1KO), and cardiomyocyte-specific Peli1 overexpressing (alpha MHC (αMHC) Peli1; AMPEL1Tg/+) animals were divided into sham and cecal ligation and puncture (CLP) surgical procedure groups. Cardiac function was determined by two-dimensional echocardiography pre-surgery and at 6- and 24-h post-surgery. Serum IL-6 and TNF-alpha levels (ELISA) (6 h), cardiac apoptosis (TUNEL assay), and Bax expression (24 h) post-surgery were measured. Results are expressed as mean ± S.E.M. RESULTS AMPEL1Tg/+ prevents sepsis-induced cardiac dysfunction assessed by echocardiographic analysis, whereas global and cardiomyocyte-specific deletion of Peli1 shows significant deterioration of cardiac functions. Cardiac function was similar across the sham groups in all three genetically modified mice. ELISA assay displayed how Peli 1 overexpression decreased cardo-suppressive circulating inflammatory cytokines (TNF-alpha, IL-6) compared to both the knockout groups. The proportion of TUNEL-positive cells varied according to Peli1 expression, with overexpression (AMPEL1Tg/+) leading to a significant reduction and Peli1 gene knockout (Peli1-/- and CP1KO) leading to a significant increase in their presence. A similar trend was also observed with Bax protein expression. The improved cellular survival associated with Peli1 overexpression was again shown with the reduction of oxidative stress marker 4-Hydroxy-2-Nonenal (4-HNE). CONCLUSION Our results indicate that overexpression of Peli1 is a novel approach that not only preserved cardiac function but reduced inflammatory markers and apoptosis following severe sepsis in a murine genetic model.
Collapse
Affiliation(s)
- Mahesh Thirunavukkarasu
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Santosh Swaminathan
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Andrew Kemerley
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Seetur R. Pradeep
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Sue Ting Lim
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Diego Accorsi
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Rickesha Wilson
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Jacob Campbell
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Ibnalwalid Saad
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - Siu-Pok Yee
- Center for Mouse Genome Modification, University of Connecticut Health School of Medicine, Farmington, CT 06032, USA
| | - J. Alexander Palesty
- Stanley J. Dudrick, Department of Surgery, Saint Mary’s Hospital, Waterbury, CT 06706, USA
| | - David W. McFadden
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Nilanjana Maulik
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT 06032, USA
- Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| |
Collapse
|
42
|
Barber G, Tanic J, Leligdowicz A. Circulating protein and lipid markers of early sepsis diagnosis and prognosis: a scoping review. Curr Opin Lipidol 2023; 34:70-81. [PMID: 36861948 DOI: 10.1097/mol.0000000000000870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
PURPOSE OF REVIEW Sepsis is the extreme response to infection associated with high mortality, yet reliable biomarkers for its identification and stratification are lacking. RECENT FINDINGS Our scoping review of studies published from January 2017 to September 2022 that investigated circulating protein and lipid markers to inform non-COVID-19 sepsis diagnosis and prognosis identified interleukin (IL)-6, IL-8, heparin-binding protein (HBP), and angiopoietin-2 as having the most evidence. Biomarkers can be grouped according to sepsis pathobiology to inform biological data interpretation and four such physiologic processes include: immune regulation, endothelial injury and coagulopathy, cellular injury, and organ injury. Relative to proteins, the pleiotropic effects of lipid species' render their categorization more difficult. Circulating lipids are relatively less well studied in sepsis, however, low high-density lipoprotein (HDL) is associated with poor outcome. SUMMARY There is a lack of robust, large, and multicenter studies to support the routine use of circulating proteins and lipids for sepsis diagnosis or prognosis. Future studies will benefit from standardizing cohort design as well as analytical and reporting strategies. Incorporating biomarker dynamic changes and clinical data in statistical modeling may improve specificity for sepsis diagnosis and prognosis. To guide future clinical decisions at the bedside, point-of-care circulating biomarker quantification is needed.
Collapse
Affiliation(s)
- Gemma Barber
- Schulich School of Medicine and Dentistry
- Robarts Research Insitute
| | | | - Aleksandra Leligdowicz
- Schulich School of Medicine and Dentistry
- Robarts Research Insitute
- Department of Medicine, Division of Critical Care, Western University, London, ON, Canada
| |
Collapse
|
43
|
Lima MR, Silva D. Septic cardiomyopathy: A narrative review. Rev Port Cardiol 2023; 42:471-481. [PMID: 36893835 DOI: 10.1016/j.repc.2021.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/03/2021] [Accepted: 05/01/2021] [Indexed: 03/09/2023] Open
Abstract
Sepsis is a systemic inflammatory response syndrome of suspected or documented infectious origin, whose outcome is multiorgan failure. Sepsis-induced myocardial dysfunction (SIMD), present in more than 50% of septic patients, is characterized by (i) left ventricular (LV) dilatation with normal or low filling pressure, (ii) right and/or LV (systolic and/or diastolic) dysfunction and (iii) reversibility. Since the first definition proposed by Parker et al. in 1984, attempts have been made to define SIMD. Many parameters are used to assess cardiac function in septic patients, sometimes making it more difficult to measure due to the intrinsic hemodynamical changes in this condition. Nevertheless, with advanced echocardiographic techniques, such as speckle tracking analysis, it is possible to diagnose and assess systolic and diastolic dysfunction, even in the earliest stages of sepsis. Cardiac magnetic resonance imaging brings new insights into the reversibility of this condition. Many uncertainties still remain regarding the mechanisms, characteristics, treatment and even prognosis of this condition. There are also inconsistent conclusions from studies, therefore this review attempts to summarize our current knowledge of SIMD.
Collapse
Affiliation(s)
- Maria Rita Lima
- Internal Medicine Department, Egas Moniz Hospital, Lisbon Ocidental Hospital Center, Lisbon, Portugal.
| | - Doroteia Silva
- Intensive Care Department, Santa Maria University Hospital, Lisbon North Hospital Center, Lisbon, Portugal; CCUL, Lisbon Academic Medical Center, Faculty of Medicine of Lisbon, Lisbon, Portugal
| |
Collapse
|
44
|
Mokhtari B, Hamidi M, Badalzadeh R, Mahmoodpoor A. Mitochondrial transplantation protects against sepsis-induced myocardial dysfunction by modulating mitochondrial biogenesis and fission/fusion and inflammatory response. Mol Biol Rep 2023; 50:2147-2158. [PMID: 36565415 DOI: 10.1007/s11033-022-08115-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/10/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Sepsis-induced myocardial dysfunction is associated with worse clinical outcomes and high mortality, but no effective therapeutic intervention has been explored, reinforcing the urgent need to develop innovative strategies. Mitochondrial dysfunction underlies the pathogenesis of sepsis-induced myocardial dysfunction. Herein, we assessed the effect of mitochondrial transplantation on sepsis-induced myocardial dysfunction in a rat model of cecal ligation and puncture (CLP)-induced sepsis. METHODS Male Wistar rats (n = 80, 12 weeks old, 250-300 g) were divided into groups with/without CLP-induced sepsis receiving mitochondrial transplantation in single or two repetitive injections (1 h or 1 and 7 h post-CLP, respectively). Mitochondria were isolated from donor rats and injected intravenously (400 µl of mitochondrial suspension containing 7.5 × 106 mitochondria/ml of respiration buffer) in recipient groups. Twenty-four hours post-operation, LDH and cTn-I levels, mitochondrial functional endpoints, expression of mitochondrial biogenesis (SIRT-1 and PGC-1α) and fission/fusion (Drp1/Mfn1 and Mfn2) genes, and inflammatory cytokines (TNF-α, IL-1β, and IL-6) levels were evaluated. Survival was tested over 72 h post-operation. RESULTS Mitotherapy significantly improved 72-hours survival (P < .05) and decreased LDH and cTn-I levels (P < .01). It also restored mitochondrial function and expression of mitochondrial biogenesis and fusion genes, and decreased the expression of mitochondrial fission gene and the levels of inflammatory cytokines (P < .05 to P < .01). Mitotherapy with repetitive injections at 1 and 7 h post-CLP provided noticeable mitoprotection in comparison with the group receiving mitotherapy at single injection. CONCLUSION Mitotherapy improved mitochondrial function, biogenesis, and dynamic associated with SIRT-1/PGC-1α network and suppressed inflammatory response in CLP-induced sepsis model, therefore, offers a promising strategy to overcome life-threatening sepsis challenge.
Collapse
Affiliation(s)
- Behnaz Mokhtari
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Hamidi
- Anesthesiology Department, Ali Nasab Hospital, Tabriz, Iran
| | - Reza Badalzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ata Mahmoodpoor
- Evidence-based Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
Abrams ST, Alhamdi Y, Zi M, Guo F, Du M, Wang G, Cartwright EJ, Toh CH. Extracellular Histone-Induced Protein Kinase C Alpha Activation and Troponin Phosphorylation Is a Potential Mechanism of Cardiac Contractility Depression in Sepsis. Int J Mol Sci 2023; 24:ijms24043225. [PMID: 36834636 PMCID: PMC9967552 DOI: 10.3390/ijms24043225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Reduction in cardiac contractility is common in severe sepsis. However, the pathological mechanism is still not fully understood. Recently it has been found that circulating histones released after extensive immune cell death play important roles in multiple organ injury and disfunction, particularly in cardiomyocyte injury and contractility reduction. How extracellular histones cause cardiac contractility depression is still not fully clear. In this work, using cultured cardiomyocytes and a histone infusion mouse model, we demonstrate that clinically relevant histone concentrations cause significant increases in intracellular calcium concentrations with subsequent activation and enriched localization of calcium-dependent protein kinase C (PKC) α and βII into the myofilament fraction of cardiomyocytes in vitro and in vivo. Furthermore, histones induced dose-dependent phosphorylation of cardiac troponin I (cTnI) at the PKC-regulated phosphorylation residues (S43 and T144) in cultured cardiomyocytes, which was also confirmed in murine cardiomyocytes following intravenous histone injection. Specific inhibitors against PKCα and PKCβII revealed that histone-induced cTnI phosphorylation was mainly mediated by PKCα activation, but not PKCβII. Blocking PKCα also significantly abrogated histone-induced deterioration in peak shortening, duration and the velocity of shortening, and re-lengthening of cardiomyocyte contractility. These in vitro and in vivo findings collectively indicate a potential mechanism of histone-induced cardiomyocyte dysfunction driven by PKCα activation with subsequent enhanced phosphorylation of cTnI. These findings also indicate a potential mechanism of clinical cardiac dysfunction in sepsis and other critical illnesses with high levels of circulating histones, which holds the potential translational benefit to these patients by targeting circulating histones and downstream pathways.
Collapse
Affiliation(s)
- Simon T. Abrams
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
| | - Yasir Alhamdi
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield S5 7AU, UK
| | - Min Zi
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Fengmei Guo
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- The Medical School, Southeast University, Nanjing 210009, China
| | - Min Du
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
| | - Guozheng Wang
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| | - Elizabeth J. Cartwright
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Roald Dahl Haemostasis & Thrombosis Centre, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| |
Collapse
|
46
|
Morris DC, Zhang ZG, Jaehne AK, Zhang J, Rivers EP. CLINICAL, MOLECULAR, AND EXOSOMAL MECHANISMS OF CARDIAC AND BRAIN DYSFUNCTION IN SEPSIS. Shock 2023; 59:173-179. [PMID: 36731014 DOI: 10.1097/shk.0000000000002015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
ABSTRACT Sepsis is a complex disease resulting from a dysregulated inflammatory response to an infection. Initiation of sepsis occurs from a localized infection that disseminates to the bloodstream placing all organ systems at risk. Septic shock is classically observed to manifest itself as systemic hypotension with hyporesponsiveness to vasopressor agents. Myocardial dysfunction occurs resulting in an inability to perfuse major organ systems throughout the body. Most importantly, the brain is hypoperfused creating an ischemic and inflammatory state resulting in the clinical observation of acute mental status changes and cognitive dysfunction commonly known as sepsis-associated encephalopathy. This short review describes the inflammatory molecular mechanisms of myocardial dysfunction, discusses the evidence of the dual roles of the microglia resulting in blood-brain barrier disruption, and suggests that septic-derived exosomes, endosome-derived lipid bilayer spheroids released from living cells, influence cardiac and neurological cellular function.
Collapse
Affiliation(s)
- Daniel C Morris
- Department of Emergency Medicine, Henry Ford Health, Detroit, Michigan
| | - Zheng Gang Zhang
- Department of Neurological Research, Henry Ford Health, Detroit, Michigan
| | - Anja K Jaehne
- Department of Emergency Medicine, Henry Ford Health, Detroit, Michigan
| | - Jing Zhang
- Department of Neurological Research, Henry Ford Health, Detroit, Michigan
| | | |
Collapse
|
47
|
DAMPs Released from Proinflammatory Macrophages Induce Inflammation in Cardiomyocytes via Activation of TLR4 and TNFR. Int J Mol Sci 2022; 23:ijms232415522. [PMID: 36555168 PMCID: PMC9778802 DOI: 10.3390/ijms232415522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiac dysfunction is a life-threatening complication in sepsis. Upon infection and cardiac stress, the cardiac macrophage population expands. Recruited macrophages exhibit a predominantly proinflammatory phenotype and release danger-associated molecular patterns (DAMPs) that contribute to cardiac dysfunction. However, the underlying pathomechanisms are highly complex and not fully understood. Here, we utilized an indirect macrophage-cardiomyocyte co-culture model to study the effects of proinflammatory macrophages on the activation of different cardiac receptors (TLR3, TLR4, and TNFR) and their role in cardiac inflammation and caspase-3/7 activation. The stimulation of cardiomyocytes with conditioned medium of LPS-stimulated macrophages resulted in elevated IL-6 protein concentrations and relative IL-6 and TNFα mRNA levels. Conditioned medium from LPS-stimulated macrophages also induced NFκB translocation and increased caspase-3/7 activation in cardiomyocytes. Analyzing the role of different cardiac receptors, we found that TLR4 and TNFR inhibition reduces cardiac inflammation and that the inhibition of TNFR prevents NFκB translocation into the nuclei of cardiomyocytes, induced by exposure to conditioned medium of proinflammatory macrophages. Moreover, we demonstrated that TLR3 inhibition reduces macrophage-mediated caspase-3/7 activation. Our results suggest that the immune response of macrophages under inflammatory conditions leads to the release of DAMPs, such as eRNA and cytokines, which in turn induce cardiomyocyte dysfunction. Thus, the data obtained in this study contribute to a better understanding of the pathophysiological mechanisms of cardiac dysfunction.
Collapse
|
48
|
Wang Z, Xiao D, Ji Q, Li Y, Cai Z, Fang L, Huo H, Zhou G, Yan X, Shen L, He B. Jujuboside A attenuates sepsis-induced cardiomyopathy by inhibiting inflammation and regulating autophagy. Eur J Pharmacol 2022; 947:175451. [PMID: 36502962 DOI: 10.1016/j.ejphar.2022.175451] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Jujuboside A (JuA), as a main effective component of Jujubogenin, has long been known as a sedative-hypnotic drug. The aim of the current study was to investigate the potential effect of JuA on sepsis-induced cardiomyopathy (SIC) induced by lipopolysaccharide (LPS). METHOD Wide type C57BL/6 mice and neonatal rat cardiomyocytes (NRCMs) were exposed to LPS to establish myocardial toxicity models. Cardiac function of septic mice was detected by echocardiography. Moreover, the survival rate was calculated for 7 days. ELISA assays were used to analyze inflammatory factors in serum. Furthermore, western blotting, flow cytometry and TUNEL staining were performed to assess cell apoptosis and transmission electron microscopy detect the number of autophagosomes in myocardium. Finally, the expression of proteins related to pyroptosis, autophagy and oxidative stress was analyzed by western blotting and immunohistochemistry staining. RESULTS Results showed that JuA pretreatment significantly improved the survival rate and cardiac function, and suppressed systemic inflammatory response in septic mice. Further study revealed that JuA could decrease cell apoptosis and pyroptosis; instead, it strengthened autophagy in SIC. Moreover, JuA also significantly decreased oxidative stress and nitrodative stress, as evidenced by suppressing the superoxide production and downregulating iNOS and gp91 expression in vivo. In addition, the autophagy inhibitor 3-MA significantly abolished the effect of JuA on autophagic activity in SIC. CONCLUSION In conclusion, the findings indicated that JuA attenuates cardiac function via blocking inflammasome-mediated apoptosis and pyroptosis, at the same time by enhancing autophagy in SIC, heralding JuA as a potential therapy for sepsis.
Collapse
|
49
|
Vivas W, Weis S. Tidy up - The unfolded protein response in sepsis. Front Immunol 2022; 13:980680. [PMID: 36341413 PMCID: PMC9632622 DOI: 10.3389/fimmu.2022.980680] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
Pathogens, their toxic byproducts, and the subsequent immune reaction exert different forms of stress and damage to the tissue of the infected host. This stress can trigger specific transcriptional and post-transcriptional programs that have evolved to limit the pathogenesis of infectious diseases by conferring tissue damage control. If these programs fail, infectious diseases can take a severe course including organ dysfunction and damage, a phenomenon that is known as sepsis and which is associated with high mortality. One of the key adaptive mechanisms to counter infection-associated stress is the unfolded protein response (UPR), aiming to reduce endoplasmic reticulum stress and restore protein homeostasis. This is mediated via a set of diverse and complementary mechanisms, i.e. the reduction of protein translation, increase of protein folding capacity, and increase of polyubiquitination of misfolded proteins and subsequent proteasomal degradation. However, UPR is not exclusively beneficial since its enhanced or prolonged activation might lead to detrimental effects such as cell death. Thus, fine-tuning and time-restricted regulation of the UPR should diminish disease severity of infectious disease and improve the outcome of sepsis while not bearing long-term consequences. In this review, we describe the current knowledge of the UPR, its role in infectious diseases, regulation mechanisms, and further clinical implications in sepsis.
Collapse
Affiliation(s)
- Wolfgang Vivas
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich Schiller University, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- *Correspondence: Wolfgang Vivas,
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich Schiller University, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich Schiller University, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
50
|
Sato K, Naito A, Shiratori T, Yamamoto M, Shimane K, Mikami M, Senda M, Kume H, Suzuki M. A case of sepsis-induced cardiomyopathy successfully treated with venoarterial extracorporeal membrane oxygenation. IJU Case Rep 2022; 6:26-29. [PMID: 36605701 PMCID: PMC9807351 DOI: 10.1002/iju5.12540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/24/2022] [Indexed: 01/04/2023] Open
Abstract
Introduction Sepsis with concomitant acute pyelonephritis, secondary to urolithiasis, is common. We report a case of sepsis-induced cardiomyopathy with acute pyelonephritis, successfully managed with venoarterial extracorporeal membrane oxygenation. Case presentation A 64-year-old woman presented with fever and disturbed consciousness. Abdominal computed tomography revealed right hydronephrosis with ipsilateral ureteral stone. Despite ureteral stent placement and antibiotic treatment, her hemodynamics worsened. She was diagnosed with sepsis-induced cardiomyopathy and underwent venoarterial extracorporeal membrane oxygenation. Her hemodynamics improved rapidly; venoarterial extracorporeal membrane oxygenation was withdrawn on postoperative day-3. She was discharged from our hospital after sufficient antibiotic treatment. Conclusion Venoarterial extracorporeal membrane oxygenation may be initiated in patients with sepsis-induced cardiomyopathy. Evaluation of left ventricular ejection fraction via echocardiography is important to determine the indication for venoarterial extracorporeal membrane oxygenation.
Collapse
Affiliation(s)
- Keigo Sato
- Department of UrologyTokyo Metropolitan Bokutoh HospitalTokyoJapan,Department of UrologyChiba Tokushukai HospitalFunabashiJapan
| | - Akihiro Naito
- Department of UrologyTokyo Metropolitan Bokutoh HospitalTokyoJapan
| | - Taichi Shiratori
- Department of UrologyTokyo Metropolitan Bokutoh HospitalTokyoJapan
| | | | - Kenichi Shimane
- Department of RheumatologyTokyo Metropolitan Bokutoh HospitalTokyoJapan
| | - Manabu Mikami
- Tertiary Emergency Medical CenterTokyo Metropolitan Bokutoh HospitalTokyoJapan,Center for Antarctic ProgramsNational Institute of Polar ResearchTachikawaJapan
| | - Mariko Senda
- Department of AnesthesiologyTokyo Metropolitan Bokutoh HospitalTokyoJapan
| | - Haruki Kume
- Department of Urology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Motofumi Suzuki
- Department of UrologyTokyo Metropolitan Bokutoh HospitalTokyoJapan
| |
Collapse
|