1
|
Liu R, Xi Y, Duan X, Zhao Y, Tian Z. Exerkine-mediated organ interactions: A new interpretation of exercise on cardiovascular function improvement. Life Sci 2025; 371:123628. [PMID: 40210118 DOI: 10.1016/j.lfs.2025.123628] [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: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Cardiovascular diseases impair the structure and function of distal organs, including the liver, skeletal muscle, kidney, and adipose tissue. Exercise stimulates the interaction between the cardiovascular system and distal organs that is important for disease rehabilitation and organ health. However, the mechanisms by which exercise improves cardiovascular function through exerkine-mediated organ crosstalk remain incompletely elucidated. We used cardiovascular, exercise, exerkines, skeletal muscle, liver, kidney, and adipose tissue as keywords to search for the relevant articles, sorted out the differences between different exercise types, summarized the functions of 17 exerkines, focused on reviewing and categorizing the molecular mechanisms of interactions between the cardiovascular system and remote organs. We also look forward to future research perspectives on exercise prevention and control of chronic metabolic diseases. The aim of this review is to provide a new theoretical basis for establishing clinical rehabilitation and exercise prescriptions for cardiovascular system diseases.
Collapse
Affiliation(s)
- Renhan Liu
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China
| | - Yue Xi
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China..
| | - Xinyan Duan
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China
| | - Yifei Zhao
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China
| | - Zhenjun Tian
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China..
| |
Collapse
|
2
|
Brito J, Rin J, Duarte C, Pereira S, Morais P, Cunha N, Ferreira D, Santos R, Rigueira J, Pinto FJ, Brito D. Association of LDL-cholesterol with prognosis in patients admitted for acutely decompensated heart failure. Rev Port Cardiol 2025; 44:191-200. [PMID: 39824246 DOI: 10.1016/j.repc.2024.09.004] [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: 11/01/2023] [Revised: 07/06/2024] [Accepted: 09/17/2024] [Indexed: 01/20/2025] Open
Abstract
INTRODUCTION AND OBJECTIVES The association of low-density lipoprotein cholesterol (LDL-C) levels and prognosis in patients with heart failure (HF) remains uncertain. This study aimed to evaluate the prognostic significance of LDL-C in patients admitted for acutely decompensated HF and establish a safety cut-off value in this population. METHODS This retrospective, observational study included 167 consecutive patients admitted for acute HF. LDL-C levels were measured on hospital admission, and patients were categorized according to their estimated cardiovascular (CV) risk. The primary endpoint was all-cause mortality at one-year, while secondary endpoints included HF hospitalizations, major thrombotic events, and net clinical benefit. RESULTS During the follow-up period, 14.4% of patients died. Higher LDL-C levels were independently associated with improved survival, with a 4-fold increase in survival probability for each 1 mg/dL increase in serum LDL-C. The minimum LDL-C value not associated with increased mortality risk was 88 mg/dL. Patients with LDL-C below this cut-off had a significantly higher risk of mortality and a tendency for higher HF hospitalization risk. The net clinical benefit endpoint was also influenced by LDL-C levels, with LDL-C below 88 mg/dL associated with an increased risk of events. CONCLUSION In patients admitted for acutely decompensated HF, higher LDL-C levels were associated with reduced risk of mortality. An LDL-C value below 88 mg/dL was associated with increased mortality, suggesting the need for a more liberal LDL-C target in this specific patient population. These findings highlight the importance of considering LDL-C levels in the management and risk assessment of patients with HF.
Collapse
Affiliation(s)
- Joana Brito
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal.
| | - João Rin
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Duarte
- Serviço de Anestesiologia, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Sara Pereira
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Pedro Morais
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Nelson Cunha
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Diogo Ferreira
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Rafael Santos
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Joana Rigueira
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Fausto J Pinto
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Dulce Brito
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
3
|
Dasinger JH, Abais-Battad JM, McCrorey MK, Van Beusecum JP. Recent advances on immunity and hypertension: the new cells on the kidney block. Am J Physiol Renal Physiol 2025; 328:F301-F315. [PMID: 39853324 DOI: 10.1152/ajprenal.00309.2024] [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: 10/30/2024] [Revised: 11/26/2024] [Accepted: 01/20/2025] [Indexed: 01/26/2025] Open
Abstract
Over the past 50 years, the contribution of the immune system has been identified in the development of hypertension and renal injury. Both human and experimental animal models of hypertension have demonstrated that innate and adaptive immune cells, along with their cytokines and chemokines, modulate blood pressure fluctuations and end organ renal damage. Numerous cell types of the innate immune system, specifically monocytes, macrophages, and dendritic cells, present antigenic peptides to T cells, promoting inflammation and the elevation of blood pressure. These T cells and other adaptive immune cells migrate to vascular and tubular cells of the kidney and promote end-organ fibrosis, damage, and ultimately hypertensive injury. Through the development of high-throughput screening, novel renal and immune cell subsets have been identified as possible contributors and regulators of renal injury and hypertension. In this review, we will consider classical immunological cells and their contribution to renal inflammation, and novel cell subsets, including renal stromal cells, that could potentially shed new light on renal injury and hypertension. Finally, we will discuss how interorgan inflammation contributes to the development of hypertension and hypertension-related multiorgan damage, and explore the clinical implications of the immunological components of renal injury and hypertension.
Collapse
Affiliation(s)
- John Henry Dasinger
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, August University, Augusta, Georgia, United States
| | - Marice K McCrorey
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Justin P Van Beusecum
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Research and Development, Ralph H. Johnson VA Healthcare System, Charleston, South Carolina, United States
| |
Collapse
|
4
|
Liu L, Huang P, Wang C, Liu Y, Gao Y, Yu K. Causal Association Between Heart Failure and Sepsis: Insights from Mendelian Randomization and Observational Studies. Clin Epidemiol 2024; 16:755-767. [PMID: 39524502 PMCID: PMC11550685 DOI: 10.2147/clep.s487118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose We aimed to identify the association between heart failure (HF) with sepsis and its mortality through Mendelian randomization (MR) and observational studies. Patients and Methods In MR study, we utilized public summary statistics from genome-wide association studies (GWAS). We conducted univariable, multivariable and network MR analyses to investigate causal relationships between HF and sepsis, and mediating roles of cytokines and growth factors. We performed an observational analysis using the MIMIC-IV database. Propensity score matching (PSM) and logistic regression models were employed to explore causal relationships between HF and sepsis, besides short-, medium-, and long-term mortality associated with sepsis. Results In univariable MR analysis, there was a causal relationship between genetically predicted HF (OR = 1.15, 95% CI = 1.02-1.29, P = 0.025) and sepsis. In multivariable and network MR analyses, βNGF was independently associated with sepsis. And it mediated 17.6% (95% CI 2.45-30.72%) of HF effect on sepsis. In the real-world observational study, acute on chronic diastolic (congestive) heart failure (DCHF) (OR = 1.59, 95% CI = 1.31-1.93, P < 0.001), acute DCHF (OR = 2.52, 95% CI = 1.61-3.95, P = 0.010), and acute diastolic heart failure (DHF) (OR = 1.52, 95% CI = 1.06-2.19, P = 0.024) after PSM were associated with occurrence of sepsis. Chronic systolic (congestive) heart failure (SCHF) was associated with increased 28-day (OR = 1.75, 95% CI = 1.06-2.91, P = 0.030), 1-year (OR = 1.80, 95% CI = 1.08-3.00, P = 0.023), and 2-year (OR = 1.86, 95% CI = 1.12-3.10, P = 0.018) mortality in sepsis. Conclusion Observational and MR analyses showed a causal relationship between HF and sepsis. Chronic SCHF was related to increased short/long-term mortality in sepsis. Our study indicated βNGF a key factor in HF-induced sepsis.
Collapse
Affiliation(s)
- Linqiong Liu
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin Medical University, Harbin, People’s Republic of China
- Departments of Critical Care Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Pengfei Huang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin Medical University, Harbin, People’s Republic of China
| | - Changsong Wang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin Medical University, Harbin, People’s Republic of China
| | - Yuxi Liu
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin Medical University, Harbin, People’s Republic of China
- Departments of Critical Care Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yan Gao
- Departments of Critical Care Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Kaijiang Yu
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
5
|
Jovanovic N, Zach V, Crocini C, Bahr LS, Forslund-Startceva SK, Franz K. A gender perspective on diet, microbiome, and sex hormone interplay in cardiovascular disease. Acta Physiol (Oxf) 2024; 240:e14228. [PMID: 39263901 DOI: 10.1111/apha.14228] [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: 03/29/2024] [Revised: 07/26/2024] [Accepted: 08/24/2024] [Indexed: 09/13/2024]
Abstract
A unique interplay between body and environment embeds and reflects host-microbiome interactions that contribute to sex-differential disease susceptibility, symptomatology, and treatment outcomes. These differences derive from individual biological factors, such as sex hormone action, sex-divergent immune processes, X-linked gene dosage effects, and epigenetics, as well as from their interaction across the lifespan. The gut microbiome is increasingly recognized as a moderator of several body systems that are thus impacted by its function and composition. In humans, biological sex components further interact with gender-specific exposures such as dietary preferences, stressors, and life experiences to form a complex whole, requiring innovative methodologies to disentangle. Here, we summarize current knowledge of the interactions among sex hormones, gut microbiota, immune system, and vascular health and their relevance for sex-differential epidemiology of cardiovascular diseases. We outline clinical implications, identify knowledge gaps, and place emphasis on required future studies to address these gaps. In addition, we provide an overview of the caveats associated with conducting cardiovascular research that require consideration of sex/gender differences. While previous work has inspected several of these components separately, here we call attention to further translational utility of a combined perspective from cardiovascular translational research, gender medicine, and microbiome systems biology.
Collapse
Affiliation(s)
- Nina Jovanovic
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Veronika Zach
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Claudia Crocini
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lina Samira Bahr
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Deutsches Herzzentrum der Charité (DHZC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sofia Kirke Forslund-Startceva
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Kristina Franz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| |
Collapse
|
6
|
Wang Y, Liu X, Wu B, Tan X, Chen L, Chu H, Zhou Z, Bao X, Xu B, Gu R. Mediation effect analysis of lipoprotein levels on BMI and cardiovascular outcomes in patients with heart failure. BMC Cardiovasc Disord 2024; 24:553. [PMID: 39395939 PMCID: PMC11470738 DOI: 10.1186/s12872-024-04155-9] [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: 05/09/2024] [Accepted: 09/02/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Among heart failure patients with obesity, the prognosis is better than those with normal weight, a phenomenon known as the obesity paradox. However, it is unclear whether lipoprotein levels play a mediating role in the machine of the obesity paradox. METHODS The study included 1663 heart failure patients hospitalized from January, 2019 through August, 2022. Kaplan-Meier survival analysis and Log-rank tests were performed for three endpoints in order to determine cumulative event-free survival. We investigated the correlation between Body Max Index (BMI) and outcomes by multifactorial Cox models. Mediation analysis was applied to study the presence and magnitude of mediation effects of triglyceride, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, apolipoprotein A1 and apolipoprotein B, with the association between BMI and endpoints. RESULTS In MACCEs, the median follow-up period was 679 days. In Cox model, compared with the underweight group, a high BMI level was significantly associated with lower all-cause mortality (HR=0.47, 95%CI 0.31~0.69, p<0.001, obese vs underweight), cardiovascular mortality (HR=0.46, 95%CI 0.30~0.73, p<0.001, obese vs underweight) and the incidence of MACCEs (HR=0.68, 95%CI 0.53~0.88, p=0.003, obese vs underweight). Mediation analysis revealed that TG was the strongest mediator between BMI and endpoints, with proportions of mediated effects of 6.6% (95%CI 2.2%~18.0%, p=0.0258, in all-cause death),7.0% (95%CI 2.3%~18.9%, p=0.0301, in cardiovascular death) and 10.2% (95%CI 3.3%~27.4%, p=0.0185, in MACCEs). CONCLUSIONS There is an "obesity paradox" in patients with heart failure, and lipoprotein levels especially triglyceride mediate the association between BMI and cardiovascular outcomes.
Collapse
Affiliation(s)
- Yi Wang
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China
| | - Xiaoli Liu
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road, Nanjing, 210008, China
| | - Baochuan Wu
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China
| | - Xi Tan
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China
| | - Lin Chen
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China
| | - Heyu Chu
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Zhongshan Road, Nanjing, 210008, China
| | - Zeyu Zhou
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China
| | - Xue Bao
- Department of Cardiology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road, Nanjing, 210008, China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China.
| | - Rong Gu
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
7
|
Dean YE, Shebl MA, Doma M, Elmezayen RW, Loayza Pintado JJ, Rouzan SS, Hassan NAIF, Yaqout YE, Tokunaga A, Anozie C, ElKoumi O, Elawady SS, Mady T, Nizam SN, Etman Y, Nizam R, Hazimeh Y, Alazmy M, Aiash H. Intestinal microbiome as a diagnostic marker of coronary artery disease: a systematic review and meta-analysis. Ann Med Surg (Lond) 2024; 86:6105-6120. [PMID: 39359774 PMCID: PMC11444608 DOI: 10.1097/ms9.0000000000002516] [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: 03/20/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The intestinal microbiome has been recently linked to several metabolic and chronic disorders, one of which is coronary artery disease (CAD). Our study aimed to analyze the intestinal microbiome of CAD patients and assess the eligibility of dysbiosis as a diagnostic marker of CAD. METHODS PubMed, Scopus, Embase, and Web of Science were searched using terms, such as 'CAD' and 'microbiome'. Only observational controlled studies were included. R version 4.2.2 was used for the analysis. RESULTS A significant association was found between the CAD group and increased Simpson and Shannon Indices compared with the control group (MD=0.04, 95% CI=0.03-0.05, and MD=0.11, 95% CI=0.01-0.22, respectively). Our analysis yielded a statistically significant association between the CAD group and increased Prevotella genus (MD=13.27, 95% CI=4.12-22.42, P-value=0.004), Catenibacterium genus (MD=0.09, 95% CI=0.09-0.10), Pseudomonas genus (MD=0.54, 95% CI=0.29-0.78, P-value), and Subdoligranulum (MD=-0.06, 95% CI=-0.06 to -0.06) compared with the control group. Another significant association was detected between the CAD group and decreased Bacteroides vulgatus and Bacteroides dorei (MD=-10.31, 95% CI=-14.78 to -5.84, P-value <0.00001). CONCLUSION Dysbiosis is an acceptable diagnostic marker of CAD. Decreased B. dorei and B. vulgatus among CAD patients suggests a protective role of these bacteria. Future clinical trials are necessary to investigate the potential benefit of supplementation of these bacteria in treating or preventing CAD.
Collapse
Affiliation(s)
- Yomna E. Dean
- Alexandria University, Faculty of Medicine, Alexandria
| | | | - Mohamed Doma
- Alexandria University, Faculty of Medicine, Alexandria
| | | | | | | | | | | | | | | | - Omar ElKoumi
- Suez Universtiy, Faculty of Medicine, Suez, Egypt
| | | | - Tamer Mady
- International American University, College of Medicine, Saint Lucia
| | | | - Yasser Etman
- Texas Health Hospital Rockwall, Director of Intensive Care Unit, Rockwall, Texas, USA
| | | | - Yusef Hazimeh
- Lebanese University
- Zahraa Hospital, University Medical Center, Lebanon
| | | | - Hani Aiash
- Suez Universtiy, Faculty of Medicine, Suez, Egypt
- SUNY Upstate Medical University, Syracuse
| |
Collapse
|
8
|
Xiao JH, Wang Y, Zhang XM, Wang WX, Zhang Q, Tang YP, Yue SJ. Intestinal permeability in human cardiovascular diseases: a systematic review and meta-analysis. Front Nutr 2024; 11:1361126. [PMID: 39086542 PMCID: PMC11289889 DOI: 10.3389/fnut.2024.1361126] [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: 01/05/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Background There is a link between cardiovascular diseases and intestinal permeability, but it is not clear. This review aimed to elucidate intestinal permeability in cardiovascular diseases by meta-analysis. Methods Multidisciplinary electronic databases were searched from the database creation to April 2023. All included studies were assessed for risk of bias according to the Joanna Briggs Institute Critical Appraisal Checklist. The heterogeneity of each study was estimated using the I2 statistic, and the data were analyzed using Review Manager 5.3 and Stata 16.0. Results In total, studies in 13 pieces of literature were included in the quantitative meta-analysis. These studies were conducted among 1,321 subjects mostly older than 48. Patients had higher levels of intestinal permeability markers (lipopolysaccharide, d-lactate, zonulin, serum diamine oxidase, lipopolysaccharide-binding protein, intestinal fatty acid binding protein, and melibiose/rhamnose) than controls (standard mean difference SMD = 1.50; 95% CI = 1.31-1.88; p < 0.00001). Similarly, lipopolysaccharide levels were higher in patients than in controls (SMD = 1.61; 95% CI = 1.02-2.21; p < 0.00001); d-lactate levels were higher in patients than in controls (SMD = 1.16; 95% CI = 0.23-2.08; p = 0.01); zonulin levels were higher in patients than in controls (SMD = 1.74; 95% CI = 1.45-2.03; p < 0.00001); serum diamine oxidase levels were higher in patients than in controls (SMD = 2.51; 95% CI = 0.29-4.73; p = 0.03). Conclusion The results of the meta-analysis verified that the intestinal barrier was damaged and intestinal permeability was increased in patients with cardiovascular diseases. These markers may become a means of the diagnosis and treatment of cardiovascular diseases. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=414296, identifier CRD42023414296.
Collapse
Affiliation(s)
- Jiang-Hong Xiao
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Yu Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Xi-Mei Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Wen-Xiao Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
9
|
Baldetti L, Gallone G, Filiberti G, Pescarmona L, Cesari A, Rizza V, Roagna E, Gurrieri D, Peveri B, Nocera L, Cianfanelli L, Marcelli G, De Lio G, Boretto P, Angelini F, Gramegna M, Pazzanese V, Sacchi S, Calvo F, Ajello S, De Ferrari GM, Frea S, Scandroglio AM. Mixed Shock Complicating Cardiogenic Shock: Frequency, Predictors, and Clinical Outcomes. Circ Heart Fail 2024; 17:e011404. [PMID: 38979611 DOI: 10.1161/circheartfailure.123.011404] [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: 11/10/2023] [Accepted: 03/11/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Patients presenting with cardiogenic shock (CS) are at risk of developing mixed shock (MS), characterized by distributive-inflammatory phenotype. However, no objective definition exists for this clinical entity. METHODS We assessed the frequency, predictors, and prognostic relevance of MS complicating CS, based on a newly proposed objective definition. MS complicating CS was defined as an objective shock state secondary to both an ongoing cardiogenic cause and a distributive-inflammatory phenotype arising at least 12 hours after the initial CS diagnosis, as substantiated by predefined longitudinal changes in hemodynamics, clinical, and laboratory parameters. RESULTS Among 213 consecutive patients admitted at 2 cardiac intensive care units with CS, 13 with inflammatory-distributive features at initial presentation were excluded, leading to a cohort of 200 patients hospitalized with pure CS (67±13 years, 96% Society of Cardiovascular Angiography and Interventions CS stage class C or higher). MS complicating CS occurred in 24.5% after 120 (29-216) hours from CS diagnosis. Lower systolic arterial pressure (P=0.043), hepatic injury (P=0.049), and suspected/definite infection (P=0.013) at CS diagnosis were independent predictors of MS development. In-hospital mortality (53.1% versus 27.8%; P=0.002) and hospital stay (21 [13-48] versus 17 [9-27] days; P=0.018) were higher in the MS cohort. At logistic multivariable analysis, MS diagnosis (odds ratio [OR], 3.00 [95% CI, 1.39-6.63]; Padj=0.006), age (OR, 1.06 [95% CI, 1.03-1.10] years; Padj<0.001), admission systolic arterial pressure <100 mm Hg (OR, 2.41 [95% CI, 1.19-4.98]; Padj=0.016), and admission serum creatinine (OR, 1.61 [95% CI, 1.19-2.26]; Padj=0.003) conferred higher odds of in-hospital death, while early temporary mechanical circulatory support was associated with lower in-hospital death (OR, 0.36 [95% CI, 0.17-0.75]; Padj=0.008). CONCLUSIONS MS complicating CS, objectively defined leveraging on longitudinal changes in distributive and inflammatory features, occurs in one-fourth of patients with CS, is predicted by markers of CS severity and inflammation at CS diagnosis, and portends higher hospital mortality.
Collapse
Affiliation(s)
- Luca Baldetti
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Guglielmo Gallone
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Gaia Filiberti
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Luca Pescarmona
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Andrea Cesari
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Vincenzo Rizza
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Edoardo Roagna
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Davide Gurrieri
- Mathematics Department, Polytechnic University of Milan, Italy (D.G.)
| | - Beatrice Peveri
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Lorenzo Nocera
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Lorenzo Cianfanelli
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Gianluca Marcelli
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Giulia De Lio
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | - Paolo Boretto
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | - Filippo Angelini
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | | | - Vittorio Pazzanese
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Stefania Sacchi
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Francesco Calvo
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Silvia Ajello
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| | - Gaetano Maria De Ferrari
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
- Department of Medical Sciences, University of Turin, Italy (G.G., L.P., E.R., L.N., G.M., G.M.D.F.)
| | - Simone Frea
- Division of Cardiology, Cardiovascular and Thoracic Department, Città della Salute e della Scienza Hospital, Turin, Italy (G.G., E.R., L.N., G.M., G.D.L., P.B., F.A., G.M.D.F., S.F.)
| | - Anna Mara Scandroglio
- Cardiac Intensive Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy (L.B., G.F., A.C., V.R., B.P., L.C., M.G., V.P., S.S., F.C., S.A., A.M.S.)
| |
Collapse
|
10
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
11
|
Perticone M, Gigliotti S, Shehaj E, Maio R, Suraci E, Miceli S, Andreozzi F, Matera G, Perticone F. Gut Permeability and Immune-Mediated Inflammation in Heart Failure. Biomedicines 2024; 12:1217. [PMID: 38927424 PMCID: PMC11200601 DOI: 10.3390/biomedicines12061217] [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: 04/26/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Heart failure (HF) is characterized by low-grade immune-mediated inflammation due to increased Toll-like receptor (TLR) expression as response to endotoxin increase and dysregulated gut barrier permeability. We investigated TLR expression and possible gut dysbiosis in HF patients compared to a control group. We enrolled 80 Caucasian HF patients and 20 controls. Low-grade immune-mediated inflammation was evaluated by TLR expression, while gut dysbiosis by the detection of zonulin and bacterial endotoxin activity in a semi-quantitative (endotoxin activity assay [EAA]) and quantitative (limulus amebocyte lysate [LAL] test) way. Compared to controls, patients with HF showed significantly higher age and blood pressure values, worse metabolic profile and kidney function, higher inflammatory biomarkers levels, and lower levels of zonulin and endotoxin activity. When dividing failing patients in those with reduced ejection fraction (HF-rEF) and those with preserved ejection fraction (HF-pEF), HF-rEF patients showed significantly higher values of inflammatory biomarkers and TLR expression than HF-pEF patients. Gut permeability biomarkers inversely correlated with the severity of HF and positively with renal function. eGFR was retained as an independent predictor of zonulin variation in all the three groups of failing patients. Present data work to extend current knowledge about the role of gut microbiota in immune-mediated inflammation in HF.
Collapse
Affiliation(s)
- Maria Perticone
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (F.A.); (F.P.)
| | - Simona Gigliotti
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (S.G.); (G.M.)
| | - Ermal Shehaj
- Cardiology and Cardiovascular Intensive Care Unit, Presidio Ospedaliero “Giovanni Paolo II” di Lamezia Terme, Azienda Sanitaria Provinciale di Catanzaro, 88046 Lamezia Terme, Italy;
| | - Raffaele Maio
- Geriatrics Unit, P.O. Germaneto, Azienda Ospedaliero-Universitaria “Renato Dulbecco”, 88100 Catanzaro, Italy; (R.M.); (S.M.)
| | - Edoardo Suraci
- Internal Medicine Unit, P.O. Pugliese-Ciaccio, Azienda Ospedaliero-Universitaria “Renato Dulbecco”, 88100 Catanzaro, Italy;
| | - Sofia Miceli
- Geriatrics Unit, P.O. Germaneto, Azienda Ospedaliero-Universitaria “Renato Dulbecco”, 88100 Catanzaro, Italy; (R.M.); (S.M.)
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (F.A.); (F.P.)
| | - Giovanni Matera
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (S.G.); (G.M.)
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy; (F.A.); (F.P.)
| |
Collapse
|
12
|
Hassan A, Luqman A, Zhang K, Ullah M, Din AU, Xiaoling L, Wang G. Impact of Probiotic Lactiplantibacillus plantarum ATCC 14917 on atherosclerotic plaque and its mechanism. World J Microbiol Biotechnol 2024; 40:198. [PMID: 38727952 DOI: 10.1007/s11274-024-04010-1] [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: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 06/22/2024]
Abstract
Atherosclerosis is viewed as not just as a problem of lipid build-up in blood vessels, but also as a chronic inflammatory disease involving both innate and acquired immunity. In atherosclerosis, the inflammation of the arterial walls is the key characteristic that significantly contributes to both the instability of plaque and the occlusion of arteries by blood clots. These events ultimately lead to stroke and acute coronary syndrome. Probiotics are living microorganisms that, when consumed in the right quantities, offer advantages for one's health. The primary objective of this study was to investigate the influence of Lactiplantibacillus plantarum ATCC 14917 (ATCC 14917) on the development of atherosclerotic plaques and its underlying mechanism in Apo lipoprotein E-knockout (Apoe-/- mice). In this study, Apoe-/- mice at approximately 8 weeks of age were randomly assigned to three groups: a Normal group that received a normal chow diet, a high fat diet group that received a gavage of PBS, and a Lactiplantibacillus plantarum ATCC 14917 group that received a high fat diet and a gavage of 0.2 ml ATCC 14917 (2 × 109 CFU/mL) per day for a duration of 12 weeks. Our strain effectively reduced the size of plaques in Apoe-/- mice by regulating the expression of inflammatory markers, immune cell markers, chemokines/chemokine receptors, and tight junction proteins (TJPs). Specifically, it decreased the levels of inflammatory markers (ICAM-1, CD-60 MCP-1, F4/80, ICAM-1, and VCAM-1) in the thoracic aorta, (Ccr7, cd11c, cd4, cd80, IL-1β, TNF-α) in the colon, and increased the activity of ROS-scavenging enzymes (SOD-1 and SOD-2). It also influenced the expression of TJPs (occludin, ZO-1, claudin-3, and MUC-3). In addition, the treatment of ATCC 14917 significantly reduced the level of lipopolysaccharide in the mesenteric adipose tissue. The findings of our study demonstrated that our strain effectively decreased the size of atherosclerotic plaques by modulating inflammation, oxidative stress, intestinal integrity, and intestinal immunity.
Collapse
Affiliation(s)
- Adil Hassan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China
| | - Ameer Luqman
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Mehtab Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Ahmad Ud Din
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Liao Xiaoling
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- JinFeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
13
|
Roszkowska P, Klimczak E, Ostrycharz E, Rączka A, Wojciechowska-Koszko I, Dybus A, Cheng YH, Yu YH, Mazgaj S, Hukowska-Szematowicz B. Small Intestinal Bacterial Overgrowth (SIBO) and Twelve Groups of Related Diseases-Current State of Knowledge. Biomedicines 2024; 12:1030. [PMID: 38790992 PMCID: PMC11117733 DOI: 10.3390/biomedicines12051030] [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: 03/24/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The human gut microbiota creates a complex microbial ecosystem, characterized by its high population density, wide diversity, and complex interactions. Any imbalance of the intestinal microbiome, whether qualitative or quantitative, may have serious consequences for human health, including small intestinal bacterial overgrowth (SIBO). SIBO is defined as an increase in the number of bacteria (103-105 CFU/mL), an alteration in the bacterial composition, or both in the small intestine. The PubMed, Science Direct, Web of Science, EMBASE, and Medline databases were searched for studies on SIBO and related diseases. These diseases were divided into 12 groups: (1) gastrointestinal disorders; (2) autoimmune disease; (3) cardiovascular system disease; (4) metabolic disease; (5) endocrine disorders; (6) nephrological disorders; (7) dermatological diseases; (8) neurological diseases (9); developmental disorders; (10) mental disorders; (11) genetic diseases; and (12) gastrointestinal cancer. The purpose of this comprehensive review is to present the current state of knowledge on the relationships between SIBO and these 12 disease groups, taking into account risk factors and the causal context. This review fills the evidence gap on SIBO and presents a biological-medical approach to the problem, clearly showing the groups and diseases having a proven relationship with SIBO, as well as indicating groups within which research should continue to be expanded.
Collapse
Affiliation(s)
- Paulina Roszkowska
- Department of Diagnostic Immunology, Pomeranian Medical University, st. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (P.R.); (I.W.-K.)
| | - Emilia Klimczak
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
| | - Ewa Ostrycharz
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
- Doctoral School, University of Szczecin, st. A. Mickiewicz 16, 71-412 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, st. Wąska 13, 71-412 Szczecin, Poland
| | - Aleksandra Rączka
- Department of Genetics, West Pomeranian University of Technology, st. Aleja Piastów 45, 70-311 Szczecin, Poland; (A.R.); (A.D.)
| | - Iwona Wojciechowska-Koszko
- Department of Diagnostic Immunology, Pomeranian Medical University, st. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (P.R.); (I.W.-K.)
| | - Andrzej Dybus
- Department of Genetics, West Pomeranian University of Technology, st. Aleja Piastów 45, 70-311 Szczecin, Poland; (A.R.); (A.D.)
| | - Yeong-Hsiang Cheng
- Department of Biotechnology and Animal Science, National Ilan University, Yilan 26047, Taiwan; (Y.-H.C.); (Y.-H.Y.)
| | - Yu-Hsiang Yu
- Department of Biotechnology and Animal Science, National Ilan University, Yilan 26047, Taiwan; (Y.-H.C.); (Y.-H.Y.)
| | - Szymon Mazgaj
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
| | - Beata Hukowska-Szematowicz
- Institute of Biology, University of Szczecin, st. Z. Felczaka 3c, 71-412 Szczecin, Poland; (E.K.); (E.O.); (S.M.)
- Molecular Biology and Biotechnology Center, University of Szczecin, st. Wąska 13, 71-412 Szczecin, Poland
| |
Collapse
|
14
|
Gale SE, Willeford A, Sandquist K, Watson K. Intravenous iron in patients with iron deficiency and heart failure: a review of modern evidence. Curr Opin Cardiol 2024; 39:178-187. [PMID: 38353280 DOI: 10.1097/hco.0000000000001121] [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] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW Iron deficiency is common in patients with heart failure, affecting up to half of ambulatory patients and an even greater percentage of patients admitted for acute decompensation. Iron deficiency in this population is also associated with poor outcomes, including worse quality of life in addition to increased hospitalizations for heart failure and mortality. Evidence suggests that patients with iron deficiency in heart failure may benefit from repletion with IV iron. RECENT FINDINGS In this review, we outline the etiology and pathophysiology of iron deficiency in heart failure as well as various iron formulations available. We discuss evidence for intravenous iron repletion with a particular focus on recent studies that have evaluated its effects on hospitalizations and mortality. Finally, we discuss areas of uncertainty and future study and provide practical guidance for iron repletion. SUMMARY In summary, there is overwhelming evidence that intravenous iron repletion in patients with iron deficiency in heart failure is both beneficial and safe. However, further evidence is needed to better identify which patients would most benefit from iron repletion as well as the ideal repletion strategy.
Collapse
Affiliation(s)
- Stormi E Gale
- Novant Health Heart and Vascular Institute, Huntersville, North Carolina
| | - Andrew Willeford
- University of California San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, San Diego, California
| | | | - Kristin Watson
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Boulet J, Sridhar VS, Bouabdallaoui N, Tardif JC, White M. Inflammation in heart failure: pathophysiology and therapeutic strategies. Inflamm Res 2024; 73:709-723. [PMID: 38546848 PMCID: PMC11058911 DOI: 10.1007/s00011-023-01845-6] [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: 09/07/2023] [Revised: 11/09/2023] [Accepted: 12/19/2023] [Indexed: 04/30/2024] Open
Abstract
A role for inflammation in the development and progression of heart failure (HF) has been proposed for decades. Multiple studies have demonstrated the potential involvement of several groups of cytokines and chemokines in acute and chronic HF, though targeting these pathways in early therapeutic trials have produced mixed results. These studies served to highlight the complexity and nuances of how pro-inflammatory pathways contribute to the pathogenesis of HF. More recent investigations have highlighted how inflammation may play distinct roles based on HF syndrome phenotypes, findings that may guide the development of novel therapies. In this review, we propose a contemporary update on the role of inflammation mediated by the innate and adaptive immune systems with HF, highlighting differences that exist across the ejection fraction spectrum. This will specifically be looked at through the lens of established and novel biomarkers of inflammation. Subsequently, we review how improvements in inflammatory pathways may mediate clinical benefits of existing guideline-directed medical therapies for HF, as well as future therapies in the pipeline targeting HF and inflammation.
Collapse
Affiliation(s)
- Jacinthe Boulet
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Vikas S Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Nadia Bouabdallaoui
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada.
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, QC, H1C 1C8, Montreal, Canada.
| |
Collapse
|
16
|
Ramos RS, Rocco IS, Viceconte M, Santo JADE, Berwanger O, Santos RHN, Kalil RAK, Jatene FB, Cavalcanti AB, Zilli AC, Pimentel WDS, Hossne Junior NA, Branco JNR, Trimer R, Evora PRB, Gomes WJ, Guizilin S. Association Between Body Mass Index, Obesity, and Clinical Outcomes Following Coronary Artery Bypass Grafting in Brazil: An Analysis of One Year of Follow-up of BYPASS Registry Patients. Braz J Cardiovasc Surg 2024; 39:e20230133. [PMID: 38569010 PMCID: PMC10987126 DOI: 10.21470/1678-9741-2023-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE To investigate the association between body mass index (BMI), obesity, clinical outcomes, and mortality following coronary artery bypass grafting (CABG) in Brazil using a large sample with one year of follow-up from the Brazilian Registry of Cardiovascular Surgeries in Adults (or BYPASS) Registry database. METHODS A multicenter cohort-study enrolled 2,589 patients submitted to isolated CABG and divided them into normal weight (BMI 20.0-24.9 kg/m2), overweight (BMI 25.0-29.9 kg/m2), and obesity (BMI > 30.0 kg/m2) groups. Inpatient postoperative outcomes included the most frequently described complications and events. Collected post-discharge outcomes included rehospitalization and mortality rates within 30 days, six months, and one year of follow-up. RESULTS Sternal wound infections (SWI) rate was higher in obese compared to normal-weight patients (relative risk [RR]=5.89, 95% confidence interval [CI]=2.37-17.82; P=0.001). Rehospitalization rates in six months after discharge were higher in obesity and overweight groups than in normal weight group (χ=6.03, P=0.049); obese patients presented a 2.2-fold increase in the risk for rehospitalization within six months compared to normal-weight patients (RR=2.16, 95% CI=1.17-4.09; P=0.045). Postoperative complications and mortality rates did not differ among groups during time periods. CONCLUSION Obesity increased the risk for SWI, leading to higher rehospitalization rates and need for surgical interventions within six months following CABG. Age, female sex, and diabetes were associated with a higher risk of mortality. The obesity paradox remains controversial since BMI may not be sufficient to assess postoperative risk in light of more complex and dynamic evaluations of body composition and physical fitness.
Collapse
Affiliation(s)
- Rodrigo Santin Ramos
- Cardiology Postgraduate Program, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Isadora Salvador Rocco
- Cardiology Postgraduate Program, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
- Cardiovascular Surgery Discipline, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Marcela Viceconte
- Cardiology Postgraduate Program, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Otavio Berwanger
- Instituto de Pesquisa - IP, Hospital do Coração -
HCor, São Paulo, São Paulo, Brazil
| | | | - Renato Abdala Karam Kalil
- Instituto de Cardiologia do Rio Grande do Sul,
Fundação Universitária de Cardiologia, Porto Alegre, Rio Grande
do Sul, Brazil
| | - Fabio B. Jatene
- Instituto de Pesquisa - IP, Hospital do Coração -
HCor, São Paulo, São Paulo, Brazil
- Cardiovascular Surgery Division, Instituto do Coração
- InCor, Hospital das Clínicas da Faculdade de Medicina da Universidade de
São Paulo - HCFMUSP, São Paulo, São Paulo, Brazil
| | | | - Alexandre Cabral Zilli
- Cardiology Postgraduate Program, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Walace de Souza Pimentel
- Cardiovascular Surgery Discipline, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Nelson Américo Hossne Junior
- Cardiovascular Surgery Discipline, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - João Nelson Rodrigues Branco
- Cardiovascular Surgery Discipline, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Renata Trimer
- Department of Physical Therapy, Universidade Federal de São
Carlos, São Carlos, São Paulo, Brazil
| | - Paulo Roberto Barbora Evora
- Department of Surgery and Anatomy, Escola de Medicina de
Riberão Preto, Universidade de São Paulo, Ribeirão Preto,
São Paulo, Brazil
| | - Walter J. Gomes
- Cardiology Postgraduate Program, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
- Cardiovascular Surgery Discipline, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Solange Guizilin
- Cardiology Postgraduate Program, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
- Cardiovascular Surgery Discipline, Escola Paulista de Medicina,
Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | | |
Collapse
|
17
|
Li C, Ng JKC, Chan GCK, Fung WWS, Lai KB, Poon PYK, Luk CCW, Chow KM, Szeto CC. Gut permeability, circulating bacterial fragments and measures of congestion in peritoneal dialysis. Clin Kidney J 2024; 17:sfae056. [PMID: 38516523 PMCID: PMC10956420 DOI: 10.1093/ckj/sfae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Indexed: 03/23/2024] Open
Abstract
Background Limited data exist on the association between gut permeability, circulating bacterial fragment and volume overload in peritoneal dialysis (PD) patients. We measured circulating bacterial fragments, N-terminal pro B-type natriuretic peptide (NT-proBNP), calprotectin and zonulin levels, and evaluate their association with the clinical outcomes in PD patients. Methods This was a single-center prospective study on 108 consecutive incident PD patients. Plasma endotoxin and bacterial DNA, and serum NT-proBNP, calprotectin and zonulin levels were measured. Primary outcomes were technique and patient survival, secondary outcomes were hospitalization data. Results There was no significant correlation between plasma endotoxin and bacterial DNA, and serum NT-proBNP, calprotectin and zonulin levels. The Homeostatic Model Assessment for Insulin Resistance (HOMA)-2β index, which represents insulin resistance, positively correlated with plasma bacterial DNA (r = 0.421, P < .001) and calprotectin levels (r = 0.362, P = .003), while serum NT-proBNP level correlated with the severity of volume overload and residual renal function. Serum NT-proBNP level was associated with technique survival even after adjusting for confounding factors [adjusted hazard ratio (aHR) 1.030, 95% confidence interval 1.009-1.051]. NT-proBNP level was also associated with patient survival by univariate analysis, but the association became insignificant after adjusting for confounding factors (aHR 1.010, P = .073). Similarly, NT-proBNP correlated with the number of hospitalizations and duration of hospitalization by univariate analysis, but the association became insignificant after adjusting for confounding factors. Conclusion There was no correlation between markers of gut permeability, circulating bacterial fragments and measures of congestion in PD patients. Bacterial fragments levels and gut permeability are both associated with insulin resistance. Serum NT-proBNP level is associated with the severity of volume overload and technique survival. Further studies are required to delineate the mechanism of high circulating bacterial fragment levels in PD patients.
Collapse
Affiliation(s)
- Chuanlei Li
- Carol & Richard Yu Peritoneal Dialysis Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong, China
- Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jack Kit-Chung Ng
- Carol & Richard Yu Peritoneal Dialysis Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Gordon Chun-Kau Chan
- Carol & Richard Yu Peritoneal Dialysis Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Winston Wing-Shing Fung
- Carol & Richard Yu Peritoneal Dialysis Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Ka-Bik Lai
- Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Peter Yam-Kau Poon
- Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Cathy Choi-Wan Luk
- Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kai-Ming Chow
- Carol & Richard Yu Peritoneal Dialysis Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Cheuk-Chun Szeto
- Carol & Richard Yu Peritoneal Dialysis Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, Shatin, Hong Kong, China
- Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
18
|
Laudani S, Godos J, Romano GL, Gozzo L, Di Domenico FM, Dominguez Azpíroz I, Martínez Diaz R, Giampieri F, Quiles JL, Battino M, Drago F, Galvano F, Grosso G. Isoflavones Effects on Vascular and Endothelial Outcomes: How Is the Gut Microbiota Involved? Pharmaceuticals (Basel) 2024; 17:236. [PMID: 38399451 PMCID: PMC10891971 DOI: 10.3390/ph17020236] [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: 12/29/2023] [Revised: 01/26/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Isoflavones are a group of (poly)phenols, also defined as phytoestrogens, with chemical structures comparable with estrogen, that exert weak estrogenic effects. These phytochemical compounds have been targeted for their proven antioxidant and protective effects. Recognizing the increasing prevalence of cardiovascular diseases (CVD), there is a growing interest in understanding the potential cardiovascular benefits associated with these phytochemical compounds. Gut microbiota may play a key role in mediating the effects of isoflavones on vascular and endothelial functions, as it is directly implicated in isoflavones metabolism. The findings from randomized clinical trials indicate that isoflavone supplementation may exert putative effects on vascular biomarkers among healthy individuals, but not among patients affected by cardiometabolic disorders. These results might be explained by the enzymatic transformation to which isoflavones are subjected by the gut microbiota, suggesting that a diverse composition of the microbiota may determine the diverse bioavailability of these compounds. Specifically, the conversion of isoflavones in equol-a microbiota-derived metabolite-seems to differ between individuals. Further studies are needed to clarify the intricate molecular mechanisms behind these contrasting results.
Collapse
Affiliation(s)
- Samuele Laudani
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.L.); (F.M.D.D.); (F.D.); (F.G.); (G.G.)
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.L.); (F.M.D.D.); (F.D.); (F.G.); (G.G.)
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy;
| | - Lucia Gozzo
- Clinical Pharmacology Unit/Regional Pharmacovigilance Centre, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-S. Marco”, 95123 Catania, Italy;
| | - Federica Martina Di Domenico
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.L.); (F.M.D.D.); (F.D.); (F.G.); (G.G.)
| | - Irma Dominguez Azpíroz
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (I.D.A.); (R.M.D.); (F.G.); (J.L.Q.); (M.B.)
- Universidade Internacional do Cuanza, Cuito EN250, Angola
- Universidad de La Romana, La Romana 22000, Dominican Republic
| | - Raquel Martínez Diaz
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (I.D.A.); (R.M.D.); (F.G.); (J.L.Q.); (M.B.)
- Universidad Internacional Iberoamericana, Campeche 24560, Mexico
- Universidad Internacional Iberoamericana, Arecibo 00613, Puerto Rico
| | - Francesca Giampieri
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (I.D.A.); (R.M.D.); (F.G.); (J.L.Q.); (M.B.)
- Department of Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy
| | - José L. Quiles
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (I.D.A.); (R.M.D.); (F.G.); (J.L.Q.); (M.B.)
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Research and Development Functional Food Centre (CIDAF), Health Science Technological Park, Avenida del Conocimiento 37, 18016 Granada, Spain
| | - Maurizio Battino
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain; (I.D.A.); (R.M.D.); (F.G.); (J.L.Q.); (M.B.)
- Department of Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang 212013, China
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.L.); (F.M.D.D.); (F.D.); (F.G.); (G.G.)
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.L.); (F.M.D.D.); (F.D.); (F.G.); (G.G.)
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (S.L.); (F.M.D.D.); (F.D.); (F.G.); (G.G.)
- Center for Human Nutrition and Mediterranean Foods (NUTREA), University of Catania, 95123 Catania, Italy
| |
Collapse
|
19
|
Song Y, Wei H, Zhou Z, Wang H, Hang W, Wu J, Wang DW. Gut microbiota-dependent phenylacetylglutamine in cardiovascular disease: current knowledge and new insights. Front Med 2024; 18:31-45. [PMID: 38424375 DOI: 10.1007/s11684-024-1055-9] [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: 08/28/2023] [Accepted: 12/04/2023] [Indexed: 03/02/2024]
Abstract
Phenylacetylglutamine (PAGln) is an amino acid derivate that comes from the amino acid phenylalanine. There are increasing studies showing that the level of PAGln is associated with the risk of different cardiovascular diseases. In this review, we discussed the metabolic pathway of PAGln production and the quantitative measurement methods of PAGln. We summarized the epidemiological evidence to show the role of PAGln in diagnostic and prognostic value in several cardiovascular diseases, such as heart failure, coronary heart disease/atherosclerosis, and cardiac arrhythmia. The underlying mechanism of PAGln is now considered to be related to the thrombotic potential of platelets via adrenergic receptors. Besides, other possible mechanisms such as inflammatory response and oxidative stress could also be induced by PAGln. Moreover, since PAGln is produced across different organs including the intestine, liver, and kidney, the cross-talk among multiple organs focused on the function of this uremic toxic metabolite. Finally, the prognostic value of PAGln compared to the classical biomarker was discussed and we also highlighted important gaps in knowledge and areas requiring future investigation of PAGln in cardiovascular diseases.
Collapse
Affiliation(s)
- Yaonan Song
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Zhitong Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Huiqing Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Weijian Hang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Junfang Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| |
Collapse
|
20
|
Kayani M, Fatima N, Yarra PC, Almansouri NE, K D, Balasubramanian A, Parvathaneni N, Mowo-Wale AG, Valdez JA, Nazir Z. Novel Biomarkers in Early Detection of Heart Failure: A Narrative Review. Cureus 2024; 16:e53445. [PMID: 38435138 PMCID: PMC10909379 DOI: 10.7759/cureus.53445] [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] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Heart failure (HF) represents a significant global health challenge, characterized by a variety of symptoms resulting from cardiac dysfunction. This dysfunction often leads to systemic and pulmonary congestion. The pathophysiology of HF is complex, involving stimulation of the sympathetic nervous system, which is insufficiently balanced by the release of natriuretic peptide. This imbalance leads to progressive hypertrophy and dilatation of the heart's chambers, impairing its pumping efficiency and increasing the risk of arrhythmias and conduction disorders. The prevalence of HF is exceptionally high in industrialized nations and is expected to increase owing to an aging population and advancements in diagnostic methods. This study emphasizes the critical role of early diagnosis in reducing morbidity and mortality associated with HF, focusing specifically on the evolving importance of biomarkers in managing this condition. Biomarkers have played a key role in transforming the diagnosis and treatment of HF. Traditional biomarkers such as b-type natriuretic peptide and N-terminal pro-b-type natriuretic peptide have been widely adopted for their cost-effectiveness and ease of access. However, the rise of novel biomarkers such as growth differentiation factor 15 and adrenomedullin has shown promising results, offering superior sensitivity and specificity. These new biomarkers enhance diagnostic accuracy, risk stratification, and prognostic evaluation in HF patients. Despite these advancements, challenges remain, such as limited availability, high costs, and the need for further validation in diverse patient populations. Through a comprehensive literature review across databases such as PubMed, Google Scholar, and the Cochrane Library, this study compiles and analyzes data from 18 relevant studies, offering a detailed understanding of the current state of HF biomarkers. The study examines both traditional and emerging biomarkers such as galectin-3 and soluble suppression of tumorigenicity 2 in HF, exploring their clinical roles and impact on patient outcomes.
Collapse
Affiliation(s)
- Maryam Kayani
- Cardiology, Shifa Tameer-e-Millat University Shifa College of Medicine, Islamabad, PAK
| | - Neha Fatima
- Internal Medicine, Lisie Hospital, Kochi, IND
| | | | - Naiela E Almansouri
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, University of Tripoli, Tripoli, LBY
| | - Deepshikha K
- Cardiology, Pondicherry Institute of Medical Sciences, Pondicherry, IND
| | | | | | | | - Josue A Valdez
- General Practice, Universidad Autónoma de Durango, Los Mochis, MEX
| | - Zahra Nazir
- Internal Medicine, Combined Military Hospital, Quetta, PAK
| |
Collapse
|
21
|
Luqman A, Hassan A, Ullah M, Naseem S, Ullah M, Zhang L, Din AU, Ullah K, Ahmad W, Wang G. Role of the intestinal microbiome and its therapeutic intervention in cardiovascular disorder. Front Immunol 2024; 15:1321395. [PMID: 38343539 PMCID: PMC10853344 DOI: 10.3389/fimmu.2024.1321395] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
The gut microbiome is a heterogeneous population of microbes comprising viruses, bacteria, fungi, and protozoa. Such a microbiome is essential for sustaining host equilibrium, and its impact on human health can be altered by a variety of factors such as external variables, social behavior, age, nutrition, and genetics. Gut microbes' imbalances are related to a variety of chronic diseases including cancer, obesity, and digestive disorders. Globally, recent findings show that intestinal microbes have a significant role in the formation of cardiovascular disease (CVD), which is still the primary cause of fatalities. Atherosclerosis, hypertension, diabetes, inflammation, and some inherited variables are all cardiovascular risk variables. However, studies found correlations between metabolism, intestinal flora, and dietary intake. Variations in the diversity of gut microbes and changes in their activity are thought to influence CVD etiology. Furthermore, the gut microbiota acts as an endocrine organ, producing bioactive metabolites such as TMA (trimethylamine)/TMAO (trimethylamine N-oxide), SCFA (short-chain fatty acids), and bile acids, which have a substantial impact on host wellness and disease by multiple mechanisms. The purpose of this overview is to compile current evidence highlighting the intricate links between gut microbiota, metabolites, and the development of CVD. It focuses on how intestinal dysbiosis promotes CVD risk factors such as heart failure, hypertension, and atherosclerosis. This review explores the normal physiology of intestinal microbes and potential techniques for targeting gut bacteria for CVD treatment using various microbial metabolites. It also examines the significance of gut bacteria in disease treatment, including supplements, prebiotics, probiotics, antibiotic therapies, and fecal transplantation, which is an innovative approach to the management of CVD. As a result, gut bacteria and metabolic pathways become increasingly attractive as potential targets for CVD intervention.
Collapse
Affiliation(s)
- Ameer Luqman
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| | - Adil Hassan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China
| | - Mehtab Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Mehraj Ullah
- School of Fermentation Engineering Tianjin University of Science and Technology, Tianjin, China
| | | | - Ahmad Ud Din
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, United States
| | - Kamran Ullah
- Department of Biology, The University of Haripur, Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Waqar Ahmad
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| |
Collapse
|
22
|
Usman I, Anwar A, Shukla S, Pathak P. Mechanistic Review on the Role of Gut Microbiota in the Pathology of Cardiovascular Diseases. Cardiovasc Hematol Disord Drug Targets 2024; 24:13-39. [PMID: 38879769 DOI: 10.2174/011871529x310857240607103028] [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: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 07/31/2024]
Abstract
Cardiovascular diseases (CVDs), which stand as the primary contributors to illness and death on a global scale, include vital risk factors like hyperlipidemia, hypertension, diabetes, and smoking, to name a few. However, conventional cardiovascular risk factors offer only partial insight into the complexity of CVDs. Lately, a growing body of research has illuminated that the gut microbiome and its by-products are also of paramount importance in the initiation and progression of CVDs. The gastrointestinal tract houses trillions of microorganisms, commonly known as gut microbiota, that metabolize nutrients, yielding substances like trimethylamine-N-oxide (TMAO), bile acids (BAs), short-chain fatty acids (SCFAs), indoxyl sulfate (IS), and so on. Strategies aimed at addressing these microbes and their correlated biological pathways have shown promise in the management and diagnosis of CVDs. This review offers a comprehensive examination of how the gut microbiota contributes to the pathogenesis of CVDs, particularly atherosclerosis, hypertension, heart failure (HF), and atrial fibrillation (AF), explores potential underlying mechanisms, and highlights emerging therapeutic prospects in this dynamic domain.
Collapse
Affiliation(s)
- Iqra Usman
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Aamir Anwar
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Shivang Shukla
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Priya Pathak
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| |
Collapse
|
23
|
Chen Z, Lin Y, Wang J, Yao K, Xie Y, Chen X, Zhou T. Relationship between Compound α-Ketoacid and Microinflammation in Patients with Chronic Kidney Disease. Curr Pharm Des 2024; 30:589-596. [PMID: 38477209 DOI: 10.2174/0113816128291248240131102709] [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: 11/04/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024]
Abstract
Chronic kidney disease (CKD) refers to the presence of structural or functional abnormalities in the kidneys that affect health, lasting for more than 3 months. CKD is not only the direct cause of global incidence rate and mortality, but also an important risk factor for cardiovascular disease. Persistent microinflammatory state has been recognized as an important component of CKD, which can lead to renal fibrosis and loss of renal function, and plays a crucial role in the pathophysiology and progression of the disease. Simultaneously, compound α-Ketoacid can bind nitrogen-containing metabolites in the blood and accelerate their excretion from the body, thereby reducing the level of metabolic waste, alleviating gastrointestinal reactions in patients, and reducing the inflammatory response and oxidative stress state of the body. Compound α-Ketoacid contains amino acids required by CKD patients. In this review, we explore the relationship between compound α-Ketoacid and microinflammation in patients with CKD. The review indicated that compound α-Ketoacid can improve the microinflammatory state in CKD patients by improving the nutritional status of CKD patients, improving patient's acid-base balance disorder, regulating oxidative stress, improving gut microbiota, and regulating abnormal lipid metabolism.
Collapse
Affiliation(s)
- Zaobin Chen
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yongda Lin
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jiali Wang
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Kaijin Yao
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yina Xie
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Xiutian Chen
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Tianbiao Zhou
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
24
|
Lee DU, Bahadur A, Ponder R, Lee KJ, Fan GH, Chou H, Lominadze Z. The causes of death in patients with nonalcoholic steatohepatitis following liver transplantation stratified using pre-liver transplant BMI. Hepatol Int 2023; 17:1393-1415. [PMID: 37160862 PMCID: PMC10767727 DOI: 10.1007/s12072-023-10529-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/18/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND & AIMS Determining the effects of pre-liver transplant (LT) BMI independent of underlying ascites on the post-LT outcomes of patients with nonalcoholic steatohepatitis (NASH) is needed to clarify the paradoxical and protective effects of obesity on post-LT endpoints. In order to accomplish this, we used graded severities of ascites to stratify the NASH-LT population and to perform an ascites-specific strata analysis with differing pre-LT BMI levels. METHODS 2005-2019 United Network for Organ Sharing (UNOS) Standard Transplant Analysis and Research (STAR) database was queried to select patients with NASH, who were categorized into specific sets of ascites severity: no ascites (n = 1188), mild ascites (n = 4463), and moderate ascites (n = 3525). Then, BMI classification (underweight: < 18.5, normal: 18.5-25, overweight: 25-30, obese: ≥ 30 kg/m2) was used to stratify each ascites-specific group and to compare to the post-LT mortality endpoints. Those under 18 years old and those who received living/multi-organ transplants were excluded. RESULTS Among each ascites category, there were the following numbers of normal, underweight, overweight, and obese BMI patients respectively; no ascites: 161, 4, 359, 664; mild ascites: 643, 28, 1311, 2481; and moderate ascites: 529, 25, 1030, 1941. The obese BMI cohort was at a lower risk of all-cause mortality compared to recipients with normal BMI with mild ascites (aHR: 0.79, 95% Confidence Interval (CI) 0.65-0.94, p-value = 0.010; case-incidence 47.10 vs 56.81 deaths per 1000 person-years) and moderate ascites (aHR: 0.77, 95% CI 0.63-0.94, p-value = 0.009; case-incidence 53.71 vs 66.17 deaths per 1000 person-years). In addition, the overweight BMI cohort with mild ascites demonstrated a lower hazard of all-cause mortality (aHR: 0.80, 95% CI 0.66-0.97, p-value = 0.03; case-incidence 49.09 vs 56.81 deaths per 1000 person-years). There was no difference in graft failure for the three BMI groups (underweight, overweight, and obese) in comparison to normal BMI. Furthermore, the overweight BMI group with mild ascites cohort demonstrated a lower hazard of death due to general infectious causes (aHR: 0.51, 95% CI 0.32-0.83, p = 0.006; case-incidence 6.12 vs 11.91 deaths per 1000 person-years) and sepsis (aHR: 0.49, 95% CI 0.27-0.86, p = 0.01; case-incidence 4.31 vs 8.50 deaths per 1000 person-years). CONCLUSION The paradoxical effects of obesity in reducing the risks of all-cause death appears to be in part modulated by ascites. The current study emphasizes the need to evaluate BMI with concomitant ascites severity pre-LT to accurately prognosticate post-LT outcomes when evaluating NASH patients with advanced liver disease.
Collapse
Affiliation(s)
- David Uihwan Lee
- Division of Gastroenterology and Hepatology, University of Maryland, 620 W Lexington St, Baltimore, MD, 21201, USA.
- Liver Center, Division of Gastroenterology and Hepatology, University of Maryland Medical Center, 22 S Greene St, Baltimore, MD, 21201, USA.
| | - Aneesh Bahadur
- Department of Medicine, Tufts University School of Medicine, Washington St, Boston, MA, 02111, USA
| | - Reid Ponder
- Department of Medicine, Tufts University School of Medicine, Washington St, Boston, MA, 02111, USA
| | - Ki Jung Lee
- Department of Medicine, Tufts University School of Medicine, Washington St, Boston, MA, 02111, USA
| | - Gregory Hongyuan Fan
- Department of Medicine, Tufts University School of Medicine, Washington St, Boston, MA, 02111, USA
| | - Harrison Chou
- Department of Medicine, Tufts University School of Medicine, Washington St, Boston, MA, 02111, USA
| | - Zurabi Lominadze
- Division of Gastroenterology and Hepatology, University of Maryland, 620 W Lexington St, Baltimore, MD, 21201, USA
| |
Collapse
|
25
|
Klobučar I, Hinteregger H, Lechleitner M, Trbušić M, Pregartner G, Berghold A, Sattler W, Frank S, Degoricija V. Association between Serum Free Fatty Acids and Clinical and Laboratory Parameters in Acute Heart Failure Patients. Biomedicines 2023; 11:3197. [PMID: 38137418 PMCID: PMC10740773 DOI: 10.3390/biomedicines11123197] [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: 10/19/2023] [Revised: 11/14/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Very little is known about the association between individual serum free fatty acids (FFAs) and clinical and laboratory parameters (indicators of heart failure severity) in acute heart failure (AHF) patients. Here, the baseline serum levels of FFAs, 16:0 (palmitic acid), 16:1 (palmitoleic acid), 18:0 (stearic acid), 18:1 (oleic acid), 18:2 (linoleic acid), 18:3 (alpha-linolenic acid or gamma-linolenic acid), 20:4 (arachidonic acid), 20:5 (eicosapentaenoic acid), and 22:6 (docosahexaenoic acid), were determined in 304 AHF patients (94.7% belonged to New York Heart Association functional class IV) using gas chromatography. Spearman correlation coefficients were used to examine the associations between the individual and total (the sum of all FFAs) FFAs and clinical and laboratory parameters. After applying a Bonferroni correction to correct for multiple testing, the total FFAs, as well as the individual FFAs (except FFAs 18:0, 20:5, and 22:6), were found to be significantly positively correlated with serum albumin. Only a few additional associations were found: FFA 16:0 was significantly negatively correlated with systolic pulmonary artery pressure, FFA 18:3 was significantly negatively correlated with C-reactive protein and body mass index, and FFA 20:4 was significantly negatively correlated with blood urea nitrogen. Based on our results, we conclude that in patients with severe AHF, individual and total serum FFAs are slightly associated with established laboratory and clinical parameters, which are indicators of heart failure severity.
Collapse
Affiliation(s)
- Iva Klobučar
- Department of Cardiology, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia; (I.K.); (M.T.)
| | - Helga Hinteregger
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (M.L.); (W.S.)
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (M.L.); (W.S.)
| | - Matias Trbušić
- Department of Cardiology, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia; (I.K.); (M.T.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036 Graz, Austria; (G.P.); (A.B.)
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036 Graz, Austria; (G.P.); (A.B.)
| | - Wolfgang Sattler
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (M.L.); (W.S.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (H.H.); (M.L.); (W.S.)
- BioTechMed-Graz, 8010 Graz, Austria
| | - Vesna Degoricija
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
- Department of Medicine, Sisters of Charity University Hospital Centre, 10000 Zagreb, Croatia
| |
Collapse
|
26
|
Kitzerow O, Suder P, Shukry M, Lisco SJ, Zucker IH, Wang HJ. Systemic mapping of organ plasma extravasation at multiple stages of chronic heart failure. Front Physiol 2023; 14:1288907. [PMID: 38033338 PMCID: PMC10687360 DOI: 10.3389/fphys.2023.1288907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction: Chronic Heart failure (CHF) is a highly prevalent disease that leads to significant morbidity and mortality. Diffuse vasculopathy is a commonmorbidity associated with CHF. Increased vascular permeability leading to plasma extravasation (PEx) occurs in surrounding tissues following endothelial dysfunction. Such micro- and macrovascular complications develop over time and lead to edema, inflammation, and multi-organ dysfunction in CHF. However, a systemic examination of PEx in vital organs among different time windows of CHF has never been performed. In the present study, we investigated time-dependent PEx in several major visceral organs including heart, lung, liver, spleen, kidney, duodenum, ileum, cecum, and pancreas between sham-operated and CHF rats induced by myocardial infarction (MI). Methods: Plasma extravasation was determined by colorimetric evaluation of Evans Blue (EB) concentrations at 3 days, ∼10 weeks and 4 months following MI. Results: Data show that cardiac PEx was initially high at day 3 post MI and then gradually decreased but remained at a moderately high level at ∼10 weeks and 4 months post MI. Lung PEx began at day 3 and remained significantly elevated at both ∼10 weeks and 4 months post MI. Spleen PExwas significantly increased at ∼10 weeks and 4 months but not on day 3 post MI. Liver PEx occurred early at day 3 and remain significantly increased at ∼10 weeks and 4 months post MI. For the gastrointestinal (GI) organs including duodenum, ileum and cecum, there was a general trend that PEx level gradually increased following MI and reached statistical significance at either 10 weeks or 4 months post MI. Similar to GI PEx, renal PEx was significantly elevated at 4 months post MI. Discussion: In summary, we found that MI generally incites a timedependent PEx of multiple visceral organs. However, the PEx time window for individual organs in response to the MI challenge was different, suggesting that different mechanisms are involved in the pathogenesis of PEx in these vital organs during the development of CHF.
Collapse
Affiliation(s)
- Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paul Suder
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mohanad Shukry
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Steven J. Lisco
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Han-Jun Wang
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
27
|
Hones KM, Hao KA, Cueto RJ, Wright JO, King JJ, Wright TW, Friedman RJ, Schoch BS. The Obesity Paradox: A Nonlinear Relationship Between 30-Day Postoperative Complications and Body Mass Index After Total Shoulder Arthroplasty. J Am Acad Orthop Surg 2023; 31:1165-1172. [PMID: 37656955 DOI: 10.5435/jaaos-d-23-00122] [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: 02/04/2023] [Accepted: 07/24/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND An inverse relationship coined the "obesity paradox" has been propositioned, in which body mass index (BMI) may be contradictorily protective in patients undergoing surgery or treatment of chronic disease. This study sought to investigate the BMI associated with the lowest rate of medical complications after total shoulder arthroplasty (TSA). METHODS The American College of Surgeons National Surgical Quality Improvement Project database was queried to identify adults who underwent elective primary TSA between January 2012 and December 2020. Thirty-day postoperative medical complications were extracted, which included death, readmission, pneumonia, pulmonary embolism, renal failure, and cardiac arrest, among others. BMI was classified into five categories (underweight [BMI <18.5 kg/m 2 ], normal weight [BMI ≥18.5 and <25 kg/m 2 ], overweight [BMI ≥25 and <30 kg/m 2 ], obese [BMI ≥30 and <40 kg/m 2 ], and morbidly obese [BMI ≥40 kg/m 2 ]). We examined the risk of any 30-day postoperative complications and BMI categorically and on a continuous basis using multivariable logistic regression controlling for age, sex, procedure year, and comorbidities. RESULTS Of the 31,755 TSAs, 84% were White, 56% were female, and the average age of patients was 69.2 ± 9.3 years. Thirty-day postoperative medical complications occurred in 4.53% (n = 1,440). When assessed on a continuous basis, the lowest risk was in patients with a BMI between 30 and 35 kg/m 2 . Underweight individuals (BMI <18.5 kg/m 2 ) had the highest postoperative complication rates overall. The probability of medical complications increased with age and was greater for female patients. CONCLUSION The relationship between BMI and complication risk in TSA is nonlinear. A BMI between 30 and 35 kg/m 2 was associated with the lowest risk of medical complications after TSA, and BMI<18.5 kg/m 2 had the highest risk overall, indicating some protective aspects of BMI against 30-day medical complications. Thus, obesity alone should not preclude patients from TSA eligibility, rather surgical candidacy should be evaluated in the context of patients' overall health and likelihood of benefit from TSA. LEVEL OF EVIDENCE III, Retrospective Comparative Study.
Collapse
Affiliation(s)
- Keegan M Hones
- From the College of Medicine, University of Florida, Gainesville, FL (Hones, Hao, and Cueto), the Department of Orthopaedic Surgery & Sports Medicine, University of Florida, Gainesville, FL (Jonathan O. Wright, King, and Thomas W. Wright), the Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, Charleston, SC (Friedman), and the Department of Orthopaedic Surgery, Mayo Clinic, Jacksonville, FL (Schoch)
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Mangner N, Winzer EB, Linke A, Adams V. Locomotor and respiratory muscle abnormalities in HFrEF and HFpEF. Front Cardiovasc Med 2023; 10:1149065. [PMID: 37965088 PMCID: PMC10641491 DOI: 10.3389/fcvm.2023.1149065] [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: 01/20/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Heart failure (HF) is a chronic and progressive syndrome affecting worldwide billions of patients. Exercise intolerance and early fatigue are hallmarks of HF patients either with a reduced (HFrEF) or a preserved (HFpEF) ejection fraction. Alterations of the skeletal muscle contribute to exercise intolerance in HF. This review will provide a contemporary summary of the clinical and molecular alterations currently known to occur in the skeletal muscles of both HFrEF and HFpEF, and thereby differentiate the effects on locomotor and respiratory muscles, in particular the diaphragm. Moreover, current and future therapeutic options to address skeletal muscle weakness will be discussed focusing mainly on the effects of exercise training.
Collapse
Affiliation(s)
- Norman Mangner
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ephraim B. Winzer
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| |
Collapse
|
29
|
Qi B, Yang ZJ, Huang N, Zheng WB, Gui C. Searching for Intrinsic Causality between Colonic Dysbiosis and Non-Ischemic Cardiomyopathy: A Mendelian Randomization-Based Analysis. J Cardiovasc Dev Dis 2023; 10:420. [PMID: 37887867 PMCID: PMC10607801 DOI: 10.3390/jcdd10100420] [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: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 10/28/2023] Open
Abstract
Objective: Little is known about gut microbiota (GM) and cardiomyopathy. Their causal relationship was explored using two-sample Mendelian randomization (TSMR) performed by R software. Methods: The single nucleotide polymorphisms (SNPs) were further screened based on the genome-wide association studies (GWAS) of gut microbiota and cardiomyopathy obtained from an open database. TSMR was performed using an MR-Egger regression, simple estimator based on mode, weighted median method, inverse variance weighted (IVW), weighted estimator and CML-MA-BIC to explore the causal association. And the sensitivity analysis was carried out using an MR-Egger regression and the leave-one-out sensitivity test. Results: As for 211 GM taxa, IVW results confirmed that the class Actinobacteria (OR = 0.81, p = 0.021) and genus Coprobacter (OR = 0.85, p = 0.033) were protective factors for cardiomyopathy. The phylum Firmicutes (OR = 0.87, p < 0.01), family Acidaminococcaceae (OR = 0.89, p < 0.01), genus Desulfovibrio (OR = 0.92, p = 0.030) and genus Prevotella9 (OR = 0.93, p = 0.029) were protective factors for ischemic cardiomyopathy. The family Rhodospirillaceae (OR = 1.06, p = 0.036) and genus Turicibacter (OR = 1.09, p = 0.019) were risk factors for ischemic cardiomyopathy. The genus Olsenella (OR = 0.91, p = 0.032) was a protective factor for non-ischemic cardiomyopathy. The order Rhodospirillales (OR = 1.14, p = 0.024), family Rikenellaceae (OR = 1.21, p = 0.012) and genus Gordonibacter (OR = 1.12, p = 0.019) were risk factors for non-ischemic cardiomyopathy. The robustness of MR results was reflected in the heterogeneity (p > 0.05) and pleiotropy (p > 0.05) analyses. Conclusions: A potential causal relationship of cardiomyopathy with some GM taxa has been confirmed in the current study.
Collapse
Affiliation(s)
- Bin Qi
- Department of Cardiology, The First Affliated Hospital of Guangxi Medical University, Nanning 530021, China; (B.Q.); (Z.-J.Y.); (N.H.); (W.-B.Z.)
- Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention, Nanning 530021, China
- Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning 530021, China
| | - Zhi-Jie Yang
- Department of Cardiology, The First Affliated Hospital of Guangxi Medical University, Nanning 530021, China; (B.Q.); (Z.-J.Y.); (N.H.); (W.-B.Z.)
- Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention, Nanning 530021, China
- Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning 530021, China
| | - Nan Huang
- Department of Cardiology, The First Affliated Hospital of Guangxi Medical University, Nanning 530021, China; (B.Q.); (Z.-J.Y.); (N.H.); (W.-B.Z.)
- Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention, Nanning 530021, China
- Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning 530021, China
| | - Wen-Bo Zheng
- Department of Cardiology, The First Affliated Hospital of Guangxi Medical University, Nanning 530021, China; (B.Q.); (Z.-J.Y.); (N.H.); (W.-B.Z.)
- Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention, Nanning 530021, China
- Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning 530021, China
| | - Chun Gui
- Department of Cardiology, The First Affliated Hospital of Guangxi Medical University, Nanning 530021, China; (B.Q.); (Z.-J.Y.); (N.H.); (W.-B.Z.)
- Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention, Nanning 530021, China
- Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning 530021, China
| |
Collapse
|
30
|
Zhang F, Deng S, Zhang J, Xu W, Xian D, Wang Y, Zhao Q, Liu Y, Zhu X, Peng M, Zhang L. Causality between heart failure and epigenetic age: a bidirectional Mendelian randomization study. ESC Heart Fail 2023; 10:2903-2913. [PMID: 37452462 PMCID: PMC10567637 DOI: 10.1002/ehf2.14446] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/01/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
AIMS Heart failure (HF) is a prevalent age-related cardiovascular disease with poor prognosis in the elderly population. This study aimed to establish the causal relationship between ageing and HF by conducting a bidirectional Mendelian randomization (MR) analysis on epigenetic age (a marker of ageing) and HF. METHODS AND RESULTS Genome-wide association study data for epigenetic age (GrimAge, HorvathAge, HannumAge, and PhenoAge) and HF were collected and assessed for significant genetic variables. A bidirectional MR analysis was carried out using the random-effects inverse-variance weighted (IVW) method as the primary approach, while other methods (MR-Egger, weighted median, simple mode, and weighted mode) and multiple sensitivity analyses (heterogeneity analysis, leave-one-out sensitivity analysis, and horizontal pleiotropy analysis) were employed to evaluate the impact of epigenetic age on HF and vice versa. Bidirectional MR analysis of two samples revealed that the epigenetic PhenoAge clock increased the risk of HF [IVW odds ratio (OR) 1.015, 95% confidence interval (CI) 1.002-1.028, P = 0.028 and weighted median OR 1.020, 95% CI 1.001-1.038, P = 0.039]. Other results were not statistically significant. CONCLUSIONS The bidirectional MR analysis demonstrated a causal link between genetically predicted epigenetic age and HF in individuals of European descent. Further research into epigenetic age in other populations and additional genetic information related to HF is warranted.
Collapse
Affiliation(s)
- Fengjun Zhang
- College of Acupuncture and MassageShandong University of Traditional Chinese MedicineJinanChina
| | - Shanshan Deng
- Non‐Coding RNA and Drug Discovery Key Laboratory of Sichuan ProvinceChengdu Medical CollegeChengduChina
- School of Basic Medical SciencesChengdu Medical CollegeChengduChina
| | - Jing Zhang
- Department of PediatricsShandong Second Provincial General HospitalJinanChina
| | - Wenchang Xu
- College of Acupuncture and MassageShandong University of Traditional Chinese MedicineJinanChina
| | - Dexian Xian
- College of Acupuncture and MassageShandong University of Traditional Chinese MedicineJinanChina
| | - Yuxuan Wang
- College of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Qiong Zhao
- Department of Traditional Chinese MedicineShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Yuan Liu
- Department of Traditional Chinese MedicineShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xiuli Zhu
- Department of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Min Peng
- Department of Traditional Chinese MedicineShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lin Zhang
- Department of Clinical Pharmacy, Shaoxing People's Hospital, Shaoxing HospitalZhejiang University School of MedicineShaoxingChina
| |
Collapse
|
31
|
Paraskevaidis I, Xanthopoulos A, Tsougos E, Triposkiadis F. Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ. Biomedicines 2023; 11:2574. [PMID: 37761015 PMCID: PMC10526035 DOI: 10.3390/biomedicines11092574] [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: 08/01/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
There is a bidirectional relationship between the heart and the gut. The gut microbiota, the community of gut micro-organisms themselves, is an excellent gut-homeostasis keeper since it controls the growth of potentially harmful bacteria and protects the microbiota environment. There is evidence suggesting that a diet rich in fatty acids can be metabolized and converted by gut microbiota and hepatic enzymes to trimethyl-amine N-oxide (TMAO), a product that is associated with atherogenesis, platelet dysfunction, thrombotic events, coronary artery disease, stroke, heart failure (HF), and, ultimately, death. HF, by inducing gut ischemia, congestion, and, consequently, gut barrier dysfunction, promotes the intestinal leaking of micro-organisms and their products, facilitating their entrance into circulation and thus stimulating a low-grade inflammation associated with an immune response. Drugs used for HF may alter the gut microbiota, and, conversely, gut microbiota may modify the pharmacokinetic properties of the drugs. The modification of lifestyle based mainly on exercise and a Mediterranean diet, along with the use of pre- or probiotics, may be beneficial for the gut microbiota environment. The potential role of gut microbiota in HF development and progression is the subject of this review.
Collapse
Affiliation(s)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece
| | - Filippos Triposkiadis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| |
Collapse
|
32
|
Modrego J, Ortega-Hernández A, Goirigolzarri J, Restrepo-Córdoba MA, Bäuerl C, Cortés-Macías E, Sánchez-González S, Esteban-Fernández A, Pérez-Villacastín J, Collado MC, Gómez-Garre D. Gut Microbiota and Derived Short-Chain Fatty Acids Are Linked to Evolution of Heart Failure Patients. Int J Mol Sci 2023; 24:13892. [PMID: 37762194 PMCID: PMC10530267 DOI: 10.3390/ijms241813892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
There is a lack of direct evidence regarding gut microbiota dysbiosis and changes in short-chain fatty acids (SCFAs) in heart failure (HF) patients. We sought to assess any association between gut microbiota composition, SCFA production, clinical parameters, and the inflammatory profile in a cohort of newly diagnosed HF patients. In this longitudinal prospective study, we enrolled eighteen newly diagnosed HF patients. At admission and after 12 months, blood samples were collected for the assessment of proinflammatory cytokines, monocyte populations, and endothelial dysfunction, and stool samples were collected for analysis of gut microbiota composition and quantification of SCFAs. Twelve months after the initial HF episode, patients demonstrated improved clinical parameters and reduced inflammatory state and endothelial dysfunction. This favorable evolution was associated with a reversal of microbiota dysbiosis, consisting of the increment of health-related bacteria, such as genus Bifidobacterium, and levels of SCFAs, mainly butyrate. Furthermore, there was a decrease in the abundance of pathogenic bacteria. In vitro, fecal samples collected after 12 months of follow-up exhibited lower inflammation than samples collected at admission. In conclusion, the favorable progression of HF patients after the initial episode was linked to the reversal of gut microbiota dysbiosis and increased SCFA production, particularly butyrate. Whether restoring butyrate levels or promoting the growth of butyrate-producing bacteria could serve as a complementary treatment for these patients deserves further studies.
Collapse
Affiliation(s)
- Javier Modrego
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Adriana Ortega-Hernández
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
| | - Josebe Goirigolzarri
- Servicio de Cardiología, Hospital Clínico de San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.G.); (M.A.R.-C.)
| | - María Alejandra Restrepo-Córdoba
- Servicio de Cardiología, Hospital Clínico de San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.G.); (M.A.R.-C.)
| | - Christine Bäuerl
- Instituto de Agroquímica y Tecnología de los Alimentos (IATA-CSIC), 46980 Paterna, Spain; (C.B.); (E.C.-M.); (M.C.C.)
| | - Erika Cortés-Macías
- Instituto de Agroquímica y Tecnología de los Alimentos (IATA-CSIC), 46980 Paterna, Spain; (C.B.); (E.C.-M.); (M.C.C.)
| | - Silvia Sánchez-González
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
| | | | - Julián Pérez-Villacastín
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Servicio de Cardiología, Hospital Clínico de San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.G.); (M.A.R.-C.)
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Fundación para la Investigación Interhospitalaria Cardiovascular, 28008 Madrid, Spain
| | - María Carmen Collado
- Instituto de Agroquímica y Tecnología de los Alimentos (IATA-CSIC), 46980 Paterna, Spain; (C.B.); (E.C.-M.); (M.C.C.)
| | - Dulcenombre Gómez-Garre
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
| |
Collapse
|
33
|
Diller GP, Lammers AE, Fischer A, Orwat S, Nienhaus K, Schmidt R, Radke RM, De-Torres-Alba F, Kaleschke G, Marschall U, Bauer UM, Roth J, Gerß J, Bormann E, Baumgartner H. Immunodeficiency is prevalent in congenital heart disease and associated with increased risk of emergency admissions and death. Eur Heart J 2023; 44:3250-3260. [PMID: 36747318 DOI: 10.1093/eurheartj/ehad029] [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: 06/10/2022] [Revised: 11/28/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023] Open
Abstract
AIMS To provide population-based data on the prevalence and clinical significance of immune deficiency syndromes (IDS) associated with congenital heart disease (CHD). METHODS AND RESULTS Utilizing administrative German Health System data the prevalence of increased susceptibility to infection (ISI) or confirmed IDS was assessed in CHD patients and compared with an age-matched non-congenital control group. Furthermore, the prognostic significance of IDS was assessed using all-cause mortality and freedom from emergency hospital admission. A total of 54 449 CHD patients were included. Of these 14 998 (27.5%) had ISI and 3034 (5.6%) had a documented IDS (compared with 2.9% of the age-matched general population). During an observation period of 394 289 patient-years, 3824 CHD patients died, and 31 017 patients experienced a combined event of all-cause mortality or emergency admission. On multivariable Cox proportional-hazard analysis, the presence of ISI [hazard ratio (HR): 2.14, P < 0.001] or documented IDS (HR: 1.77, P = 0.035) emerged as independent predictors of all-cause mortality. In addition, ISI and confirmed IDS were associated with a significantly higher risk of emergency hospital admission (P = 0.01 for both on competing risk analysis) during follow-up. CONCLUSION Limited immune competence is common in CHD patients and associated with an increased risk of morbidity and mortality. This highlights the need for structured IDS screening and collaboration with immunology specialists as immunodeficiency may be amenable to specific therapy. Furthermore, studies are required to assess whether IDS patients might benefit from intensified antibiotic shielding or tailored prophylaxis.
Collapse
Affiliation(s)
- Gerhard-Paul Diller
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
- Adult Congenital Heart Disease Unit, Royal Brompton Hospital and King's College, Sydney Street, Sw3 6NP London, UK
- National Register for Congenital Heart Disease, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Astrid Elisabeth Lammers
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
- Department of Paediatric Cardiology, University Hospital Münster Germany, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Alicia Fischer
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Stefan Orwat
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Klara Nienhaus
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Renate Schmidt
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Robert M Radke
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Fernando De-Torres-Alba
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Gerrit Kaleschke
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Ursula Marschall
- Department of Medicine and Health Services Research, BARMER Health Insurance, Lichtscheider Str., 8942285 Wuppertal, Germany
| | - Ulrike M Bauer
- National Register for Congenital Heart Disease, Augustenburger Platz 1, 13353 Berlin, Germany
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung), Oudenarder Str. 16, 13347 Berlin, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Joachim Gerß
- Department for Biostatistics, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Eike Bormann
- Department for Biostatistics, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Helmut Baumgartner
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
- National Register for Congenital Heart Disease, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
34
|
Yajima T, Yajima K. Association of extracellular water/total body water ratio with protein-energy wasting and mortality in patients on hemodialysis. Sci Rep 2023; 13:14257. [PMID: 37652929 PMCID: PMC10471676 DOI: 10.1038/s41598-023-41131-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
Bioimpedance analysis-assessed extracellular water/total body water (ECW/TBW) ratio may be a marker for mortality and poor nutritional status in hemodialysis patients. In 193 maintenance hemodialysis patients, we retrospectively investigated the relationships among ECW/TBW ratio, mortality, and protein-energy wasting (PEW). Four components-body mass index, normalized protein catabolic rate, normalized serum creatinine level, and serum albumin level-constitute the simple PEW score; this score was calculated based on the positive number of items concerning malnutrition among these four components. A score ≥ 3 indicated PEW. Patients were stratified by an ECW/TBW ratio cut-off value (0.40) and by PEW versus non-PEW status. The simple PEW score, cardiothoracic ratio, and log-transformed C-reactive protein level were independently correlated with the ECW/TBW ratio. Eighty-four patients died during follow-up (median 4.3 years). After adjustments for sex, age, hemodialysis vintage, histories of cardiovascular events and diabetes, and C-reactive protein level, a higher ECW/TBW ratio and PEW were independently related to elevated risks of all-cause death. Adding the ECW/TBW ratio to a baseline risk model including PEW significantly increased C-statistics from 0.788 to 0.835. In conclusion, the ECW/TBW ratio may be an indicator of PEW and may be a predictor of death even accounting for PEW, in hemodialysis patients.
Collapse
Affiliation(s)
- Takahiro Yajima
- Department of Nephrology, Matsunami General Hospital, Hashima Gun, Gifu, 501-6062, Japan.
| | - Kumiko Yajima
- Department of Internal Medicine, Matsunami General Hospital, Hashima Gun, Gifu, 501-6062, Japan
| |
Collapse
|
35
|
Chen A, Zhang J, Zhang Y. Gut microbiota in heart failure and related interventions. IMETA 2023; 2:e125. [PMID: 38867928 PMCID: PMC10989798 DOI: 10.1002/imt2.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/14/2024]
Abstract
Heart failure (HF) is a sophisticated syndrome with structural or functional impairment of ventricular filling or ejection of blood, either causing symptoms and signs or being asymptomatic. HF is a major global health issue affecting about 64.3 million people worldwide. The gut microbiota refers to the complex ecosystem of microorganisms, mainly bacteria, in the gut. Studies have revealed that the gut microbiota is associated with many diseases ranging from neurodegenerative diseases to inflammatory bowel disease and cardiovascular diseases. The gut hypothesis of HF suggests that low cardiac output and systemic circulation congestion would cause insufficient intestinal perfusion, leading to ischemia and intestinal barrier dysfunction. The resulting bacterial translocation would contribute to inflammation. Recent studies have refined the hypothesis that changes of metabolites in the gut microbiota have a close relationship with HF. Thus, the gut microbiota has emerged as a potential therapeutic target for HF due to both its critical role in regulating host physiology and metabolism and its pivotal role in the development of HF. This review article aims to provide an overview of the current understanding of the gut microbiota's involvement in HF, including the introduction of the gut hypothesis of HF, its association with HF progression, the potential mechanisms involved mediated by the gut microbiota metabolites, and the impact of various interventions on the gut microbiota, including dietary interventions, probiotic therapy, fecal microbiota transplantation, antibiotics, and so on. While the gut hypothesis of HF is refined with up-to-date knowledge and the gut microbiota presents a promising target for HF therapy, further research is still needed to further understand the underlying mechanisms between gut microbiota and HF, the efficacy of these interventions, and contribute to the health of HF patients.
Collapse
Affiliation(s)
- An‐Tian Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
| | - Jian Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
- Key Laboratory of Clinical Research for Cardiovascular MedicationsNational Health CommitteeBeijingChina
| | - Yuhui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
| |
Collapse
|
36
|
Belli M, Barone L, Longo S, Prandi FR, Lecis D, Mollace R, Margonato D, Muscoli S, Sergi D, Federici M, Barillà F. Gut Microbiota Composition and Cardiovascular Disease: A Potential New Therapeutic Target? Int J Mol Sci 2023; 24:11971. [PMID: 37569352 PMCID: PMC10418329 DOI: 10.3390/ijms241511971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/13/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
A great deal of evidence has revealed an important link between gut microbiota and the heart. In particular, the gut microbiota plays a key role in the onset of cardiovascular (CV) disease, including heart failure (HF). In HF, splanchnic hypoperfusion causes intestinal ischemia resulting in the translocation of bacteria and their metabolites into the blood circulation. Among these metabolites, the most important is Trimethylamine N-Oxide (TMAO), which is responsible, through various mechanisms, for pathological processes in different organs and tissues. In this review, we summarise the complex interaction between gut microbiota and CV disease, particularly with respect to HF, and the possible strategies for influencing its composition and function. Finally, we highlight the potential role of TMAO as a novel prognostic marker and a new therapeutic target for HF.
Collapse
Affiliation(s)
- Martina Belli
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lucy Barone
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Susanna Longo
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy (R.M.)
| | - Francesca Romana Prandi
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
- Division of Cardiology, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Dalgisio Lecis
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Rocco Mollace
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy (R.M.)
- Cardiovascular Department, Humanitas Gavazzeni, 24125 Bergamo, Italy
| | - Davide Margonato
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Saverio Muscoli
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Domenico Sergi
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| | - Massimo Federici
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy (R.M.)
| | - Francesco Barillà
- Division of Cardiology, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (M.B.)
| |
Collapse
|
37
|
Nakashima M, Akagi S, Ejiri K, Nakamura K, Ito H. Impact of malnutrition on prognosis in patients with pulmonary arterial hypertension. Pulm Circ 2023; 13:e12286. [PMID: 37705961 PMCID: PMC10496044 DOI: 10.1002/pul2.12286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/15/2023] Open
Abstract
Pulmonary arterial hypertension is a life-threatening disease that coexists with right heart failure. We evaluated the relationship between malnutrition and prognosis in patients with pulmonary arterial hypertension, as malnutrition is known as a prognosis determinant in chronic heart failure. We retrospectively reviewed data of patients with pulmonary arterial hypertension before treatment. The Geriatric Nutritional Risk Index, Prognostic Nutritional Index, and Controlling Nutritional Status scores on the day of diagnosis were calculated to assess the nutritional status. Clinical endpoints were defined as composite outcomes of all-cause death or lung transplantation. Eighty patients were enrolled (mean age, 50 years; 23 men). The mean pulmonary arterial pressure was 47 ± 19 mmHg, Geriatric Nutritional Risk Index was 99.9 ± 12.0, and Prognostic Nutritional Index was 46.3 ± 10.0. The median Controlling Nutritional Status score was 2 (1-4). During the median 5.5-year follow-up period, 28 composite events occurred. Kaplan-Meier analysis demonstrated significant differences in the incidence of clinical endpoints between groups divided by each median Geriatric Nutritional Risk Index, Prognostic Nutritional Index, and Controlling Nutritional Status score (p = 0.007, 0.039, and 0.010, respectively). In multivariate Cox regression analysis, clinical endpoints were significantly associated with Geriatric Nutritional Risk Index (hazard ratio: 0.953, 95% confidence interval: 0.918-0.990), Prognostic Nutritional Index (hazard ratio: 0.942, 95% confidence interval: 0.892-0.996), and Controlling Nutritional Status score (hazard ratio: 1.230, 95% confidence interval: 1.056-1.433) after adjustment for factors associated in univariate Cox regression analysis. Malnutrition at diagnosis is a useful prognostic predictor for patients with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Mitsutaka Nakashima
- Department of Cardiovascular Medicine, Okayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Kentaro Ejiri
- Department of Cardiovascular Medicine, Okayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
38
|
Gao K, Yu X, Li F, Huang Y, Liu J, Liu S, Lu L, Yang R, Wang C. Qishen granules regulate intestinal microecology to improve cardiac function in rats with heart failure. Front Microbiol 2023; 14:1202768. [PMID: 37396388 PMCID: PMC10307979 DOI: 10.3389/fmicb.2023.1202768] [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: 04/09/2023] [Accepted: 05/17/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction Qishen Granule (QSG), a clinically approved traditional Chinese medicine, has been researched for treating heart failure (HF) for many years. However, the effect of QSG on intestinal microecology remains unconfirmed. Therefore, this study aimed to elucidate the possible mechanism of QSG regulating HF in rats based on intestinal microecological changes. Methods A rat model with HF induced by myocardial infarction was prepared by left coronary artery ligation. Cardiac functions were assessed by echocardiography, pathological changes in the heart and ileum by hematoxylin-eosin (HE) and Masson staining, mitochondrial ultrastructure by transmission electron microscope, and gut microbiota by 16S rRNA sequencing. Results QSG administration improved cardiac function, tightened cardiomyocytes alignment, decreased fibrous tissue and collagen deposition, and reduced inflammatory cell infiltration. Electron microscopic observation of mitochondria revealed that QSG could arrange mitochondria neatly, reduce swelling, and improve the structural integrity of the crest. Firmicutes were the dominant component in the model group, and QSG could significantly increase the abundance of Bacteroidetes and Prevotellaceae_NK3B31_group. Furthermore, QSG significantly reduced plasma lipopolysaccharide (LPS), improved intestinal structure, and recovered barrier protection function in rats with HF. Conclusion These results demonstrated that QSG was able to improve cardiac function by regulating intestinal microecology in rats with HF, suggesting promising therapeutic targets for HF.
Collapse
Affiliation(s)
- Kuo Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fanghe Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yiran Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiali Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Siqi Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ran Yang
- Guanganmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chao Wang
- Zang-xiang Teaching and Research Department, The Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
39
|
Matsumori A. Nuclear Factor-κB is a Prime Candidate for the Diagnosis and Control of Inflammatory Cardiovascular Disease. Eur Cardiol 2023; 18:e40. [PMID: 37456770 PMCID: PMC10345985 DOI: 10.15420/ecr.2023.10] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/17/2023] [Indexed: 07/18/2023] Open
Abstract
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is responsible for the regulation of genes involved in inflammation and immune responses. NF-κB may play an important role in cardiovascular diseases (CVDs), atherosclerosis and diabetes. Several therapeutic agents used for the treatment of CVDs and diabetes, such as pimobendan and sodium-glucose cotransporter 2 inhibitors, exert anti-inflammatory effects by inhibiting NF-κB activation; anti-inflammatory therapy may have beneficial effects in CVDs and diabetes. Several pharmacological agents and natural compounds may inhibit NF-κB, and these agents alone or in combination may be used to treat various inflammatory diseases. Immunoglobulin-free light chains could be surrogate biomarkers of NF-κB activation and may be useful for evaluating the efficacy of these agents. This review discusses recent advances in our understanding of how the NF-κB signalling pathway controls inflammation, metabolism and immunity, and how improved knowledge of these pathways may lead to better diagnostics and therapeutics for various human diseases.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Institute, National Hospital Organization, Kyoto Medical Center Kyoto, Japan
| |
Collapse
|
40
|
Nendl A, Raju SC, Broch K, Mayerhofer CCK, Holm K, Halvorsen B, Lappegård KT, Moscavitch S, Hov JR, Seljeflot I, Trøseid M, Awoyemi A. Intestinal fatty acid binding protein is associated with cardiac function and gut dysbiosis in chronic heart failure. Front Cardiovasc Med 2023; 10:1160030. [PMID: 37332580 PMCID: PMC10272617 DOI: 10.3389/fcvm.2023.1160030] [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: 02/06/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Background The gut microbiota in patients with chronic heart failure (HF) is characterized by low bacterial diversity and reduced ability to synthesize beneficial metabolites. These changes may facilitate leakage of whole bacteria or bacterial products from the gut into the bloodstream, which may activate the innate immune system and contribute to the low-grade inflammation seen in HF. In this exploratory cross-sectional study, we aimed to investigate relationships between gut microbiota diversity, markers of gut barrier dysfunction, inflammatory markers, and cardiac function in chronic HF patients. Methods In total, 151 adult patients with stable HF and left ventricular ejection fraction (LVEF) < 40% were enrolled. We measured lipopolysaccharide (LPS), LPS-binding protein (LBP), intestinal fatty acid binding protein (I-FABP), and soluble cluster of differentiation 14 (sCD14) as markers of gut barrier dysfunction. N-terminal pro-B-type natriuretic peptide (NT-proBNP) level above median was used as a marker of severe HF. LVEF was measured by 2D-echocardiography. Stool samples were sequenced using 16S ribosomal RNA gene amplification. Shannon diversity index was used as a measure of microbiota diversity. Results Patients with severe HF (NT-proBNP > 895 pg/ml) had increased I-FABP (p < 0.001) and LBP (p = 0.03) levels. ROC analysis for I-FABP yielded an AUC of 0.70 (95% CI 0.61-0.79, p < 0.001) for predicting severe HF. A multivariate logistic regression model showed increasing I-FABP levels across quartiles of NT-proBNP (OR 2.09, 95% CI 1.28-3.41, p = 0.003). I-FABP was negatively correlated with Shannon diversity index (rho = -0.30, p = <0.001), and the bacterial genera Ruminococcus gauvreauii group, Bifidobacterium, Clostridium sensu stricto, and Parasutterella, which were depleted in patients with severe HF. Conclusions In patients with HF, I-FABP, a marker of enterocyte damage, is associated with HF severity and low microbial diversity as part of an altered gut microbiota composition. I-FABP may reflect dysbiosis and may be a marker of gut involvement in patients with HF.
Collapse
Affiliation(s)
- Andraž Nendl
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sajan C. Raju
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Kristian Holm
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut Tore Lappegård
- Division of Internal Medicine, Nordland Hospital, Bodø, Norway
- Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
| | - Samuel Moscavitch
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Johannes Roksund Hov
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Marius Trøseid
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Ayodeji Awoyemi
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| |
Collapse
|
41
|
Cui H, Han S, Dai Y, Xie W, Zheng R, Sun Y, Xia X, Deng X, Cao Y, Zhang M, Shang H. Gut microbiota and integrative traditional Chinese and western medicine in prevention and treatment of heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154885. [PMID: 37302262 DOI: 10.1016/j.phymed.2023.154885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Heart failure (HF) is the terminal stage of multiple cardiovascular diseases, with high mortality and morbidity. More and more studies have proved that gut microbiota may play a role in the process of HF, which is expected to become a new therapeutic target. The combination of traditional Chinese and Western medicine has vast therapeutic potential of complementation against HF. PURPOSE This manuscript expounds on the research progress of mechanisms of gut microbiota participating in the occurrence and prognosis of HF and the role of integrative traditional Chinese and Western medicine from 1987 to 2022. The combination of traditional Chinese and Western medicine in the prevention and treatment of HF from the perspective of gut microbiota has been discussed. METHODS Studies focusing on the effects and their mechanisms of gut microbiota in HF and the role of integrative traditional Chinese and Western medicine were identified and summarized, including contributions from February 1987 until August 2022. The investigation was carried out in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. We searched PubMed, Embase, Cochrane Library, CNKI, Wanfang, and VIP databases up to April 2023 by using the relevant keywords and operators. RESULTS A total of 34 articles were finally included in this review.16 RCTs and 13 basic researches, and 3 clinical research studies involving 7 relevant outcome indicators(cardiac function evaluation index, changes in gut microbiota, inflammatory factors, metabolites of gut microbiota, serum nutritional index protein, quality of life score, intestinal permeability and all-cause mortality). Compared with healthy controls, serum TNF-α and TMAO levels were significantly higher in patients with heart failure [MD = 5.77, 95%CI(4.97, 6.56), p < 0.0001; SMD = 1.92, 95%CI(1.70, 2.14), p < 0.0001]. Escherichia coli and Thick-walled bacteria increased significantly [SMD = -0.99, 95%CI(-1.38, -0.61), p < 0.0001, SMD = 2.58, 95%CI(2.23, 2.93), p < 0.0001];The number of bacteroides and lactobacillus decreased [SMD = -2.29, 95%CI(-2.54, -2.04), p < 0.0001; SMD = -1.55, 95%CI(-1.8, -1.3), p < 0.0001]. There was no difference in bifidobacterium [SMD = 0.16, 95%CI(-0.22, 0.54), p = 0.42]. In the published literature, it is not difficult to see that most of the results are studied and proved based on animal experiments or clinical trials, involving the cellular level, while the mechanism and mode of action of the molecular biology of traditional Chinese medicine are less elaborated, which is related to the characteristics of multi-components and multi-targets of traditional Chinese medicine. The above are the shortcomings of published literature, which can also be the direction of future research. CONCLUSION Heart failure patients have decreased beneficial bacteria such as Bacillus mimics and Lactobacillus in the intestinal flora and increased harmful flora like thick-walled flora. And increase the inflammatory response of the body and the expression of trimethylamine oxide (TMAO) in the serum. And The prevention and treatment of integrative traditional Chinese and Western medicine against heart failure based on gut microbiota and its metabolites is a promising research direction.
Collapse
Affiliation(s)
- Herong Cui
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanan Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wei Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaofeng Xia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaopeng Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yaru Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
42
|
Mamic P, Snyder M, Tang WHW. Gut Microbiome-Based Management of Patients With Heart Failure: JACC Review Topic of the Week. J Am Coll Cardiol 2023; 81:1729-1739. [PMID: 37100490 DOI: 10.1016/j.jacc.2023.02.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 04/28/2023]
Abstract
Despite therapeutic advances, chronic heart failure (HF) is still associated with significant risk of morbidity and mortality. The course of disease and responses to therapies vary widely among individuals with HF, highlighting the need for precision medicine approaches. Gut microbiome stands to be an important aspect of precision medicine in HF. Exploratory clinical studies have revealed shared patterns of gut microbiome dysregulation in this disease, with mechanistic animal studies providing evidence for active involvement of the gut microbiome in development and pathophysiology of HF. Deeper insights into gut microbiome-host interactions in patients with HF promise to deliver novel disease biomarkers, preventative and therapeutic targets, and improve disease risk stratification. This knowledge may enable a paradigm shift in how we care for patients with HF, and pave the path toward improved clinical outcomes through personalized HF care.
Collapse
Affiliation(s)
- Petra Mamic
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford University, Stanford, California, USA; Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, California, USA.
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, California, USA
| | - W H Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
43
|
Nguyen M, Gautier T, Masson D, Bouhemad B, Guinot PG. Endotoxemia in Acute Heart Failure and Cardiogenic Shock: Evidence, Mechanisms and Therapeutic Options. J Clin Med 2023; 12:jcm12072579. [PMID: 37048662 PMCID: PMC10094881 DOI: 10.3390/jcm12072579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Acute heart failure and cardiogenic shock are frequently occurring and deadly conditions. In patients with those conditions, endotoxemia related to gut injury and gut barrier dysfunction is usually described as a driver of organ dysfunction. Because endotoxemia might reciprocally alter cardiac function, this phenomenon has been suggested as a potent vicious cycle that worsens organ perfusion and leading to adverse outcomes. Yet, evidence beyond this phenomenon might be overlooked, and mechanisms are not fully understood. Subsequently, even though therapeutics available to reduce endotoxin load, there are no indications to treat endotoxemia during acute heart failure and cardiogenic shock. In this review, we first explore the evidence regarding endotoxemia in acute heart failure and cardiogenic shock. Then, we describe the main treatments for endotoxemia in the acute setting, and we present the challenges that remain before personalized treatments against endotoxemia can be used in patients with acute heart failure and cardiogenic shock.
Collapse
|
44
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
45
|
Ueno S, Murashima M, Ogawa R, Saito M, Ito S, Hayakawa S, Okubo T, Sagawa H, Tanaka T, Takahashi H, Matsuo Y, Mitsui A, Kimura M, Hamano T, Takiguchi S. The cisplatin-induced acute kidney injury is a novel risk factor for postoperative complications in patients with esophageal cancer: a retrospective cohort study. BMC Surg 2023; 23:67. [PMID: 36973771 PMCID: PMC10044717 DOI: 10.1186/s12893-023-01949-0] [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: 11/04/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Cisplatin-induced acute kidney injury (AKI) is common during preoperative chemotherapy for esophageal cancer. The purpose of this study was to investigate the association between AKI after preoperative chemotherapy and postoperative complications in patients with esophageal cancer. METHODS In this retrospective cohort study, we included patients who had received preoperative chemotherapy with cisplatin and underwent surgical resection for esophageal cancer under general anesthesia from January 2017 to February 2022 at an education hospital. A predictor was stage 2 or higher cisplatin-induced AKI (c-AKI) defined by the KDIGO criteria within 10 days after chemotherapy. Outcomes were postoperative complications and length of hospital stays. Associations between c-AKI and outcomes including postoperative complications and length of hospital stays were examined with logistic regression models. RESULTS Among 101 subjects, 22 developed c-AKI with full recovery of the estimated glomerular filtration (eGFR) before surgery. Demographics were not significantly different between patients with and without c-AKI. Patients with c-AKI had significantly longer hospital stays than those without c-AKI [mean (95% confidence interval (95%CI)) 27.6 days (23.3-31.9) and 43.8 days (26.5-61.2), respectively, mean difference (95%CI) 16.2 days (4.4-28.1)]. Those with c-AKI had higher C-reactive protein (CRP) levels and prolonged weight gain after surgery and before the events of interest despite having comparable eGFR trajectories after surgery. c-AKI was significantly associated with anastomotic leakage and postoperative pneumonia [odds ratios (95%CI) 4.14 (1.30-13.18) and 3.87 (1.35-11.0), respectively]. Propensity score adjustment and inverse probability weighing yielded similar results. Mediation analysis showed that a higher incidence of anastomotic leakage in patients with c-AKI was primarily mediated by CRP levels (mediation percentage 48%). CONCLUSION c-AKI after preoperative chemotherapy in esophageal cancer patients was significantly associated with the development of postoperative complications and led to a resultant longer hospital stay. Increased vascular permeability and tissue edema due to prolonged inflammation might explain the mechanisms for the higher incidence of postoperative complications.
Collapse
Affiliation(s)
- Shuhei Ueno
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Miho Murashima
- Department of Nephrology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Masaki Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Sunao Ito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Shunsuke Hayakawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Tomotaka Okubo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiroyuki Sagawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Tatsuya Tanaka
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Masahiro Kimura
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takayuki Hamano
- Department of Nephrology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, 565-0871, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| |
Collapse
|
46
|
Kilic O, Kaya HI, Secme M, Ki Li Nc M, Sevgican CI, Buber I, Dodurga Y, Si Msek O, Ergin C, Kilic ID. The effect of heart failure on gut microbial richness and diversity. Rev Port Cardiol 2023:S0870-2551(23)00120-8. [PMID: 36893840 DOI: 10.1016/j.repc.2022.06.016] [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: 03/09/2022] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 03/09/2023] Open
Abstract
INTRODUCTION With recent advances in genome sequencing technology, a large body of evidence has accumulated over the last few years linking alterations in microbiota with cardiovascular disease. In this study, we aimed to compare gut microbial composition using 16S ribosomal DNA (rDNA) sequencing techniques in patients with coronary artery disease (CAD) and stable heart failure (HF) with reduced ejection fraction and patients with CAD but with normal ejection fraction. We also studied the relationship between systemic inflammatory markers and microbial richness and diversity. METHODS A total of 40 patients (19 with HF and CAD, 21 with CAD but without HF) were included in the study. HF was defined as left ventricular ejection fraction <40%. Only stable ambulatory patients were included in the study. Gut microbiota were assessed from the participants' fecal samples. The diversity and richness of microbial populations in each sample were assessed by the Chao1-estimated OTU number and the Shannon index. RESULTS The Chao1-estimated OTU number and Shannon index were similar between HF and control groups. There was no statistically significant relationship between inflammatory marker levels (tumor necrosis factor-alpha, interleukin 1-beta, endotoxin, C-reactive protein, galectin-3, interleukin 6, and lipopolysaccharide-binding protein) and microbial richness and diversity when analyzed at the phylum level. CONCLUSION In the current study, compared to patients with CAD but without HF, stable HF patients with CAD did not show changes in gut microbial richness and diversity. At the genus level Enterococcus sp. was more commonly identified in HF patients, in addition to certain changes in species levels, including increased Lactobacillus letivazi.
Collapse
Affiliation(s)
- Oguz Kilic
- Department of Cardiology, Karaman Training and Research Hospital, Karaman, Turkey; Department of Cardiology, Pamukkale University Hospitals, Denizli, Turkey.
| | - Halil Ibrahim Kaya
- Department of Food Engineering, University of Bayburt University, Bayburt, Turkey
| | - Mucahit Secme
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Mehmet Ki Li Nc
- Department of Cardiology, Konya City Hospital, Konya, Turkey; Department of Cardiology, Pamukkale University Hospitals, Denizli, Turkey
| | | | - Ipek Buber
- Department of Cardiology, Pamukkale University Hospitals, Denizli, Turkey
| | - Yavuz Dodurga
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Omer Si Msek
- Department of Food Engineering, University of Pamukkale University, Denizli, Turkey
| | - Cagrı Ergin
- Department of Medical Microbiology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Ismail Dogu Kilic
- Department of Cardiology, Pamukkale University Hospitals, Denizli, Turkey
| |
Collapse
|
47
|
Kida K, Miyajima I, Suzuki N, Greenberg BH, Akashi YJ. Nutritional management of heart failure. J Cardiol 2023; 81:283-291. [PMID: 36370995 DOI: 10.1016/j.jjcc.2022.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022]
Abstract
Nutrition in the cardiovascular field to date has focused on improving lifestyle-related diseases such as hypertension and diabetes from the viewpoint of secondary prevention. For these conditions, "nutrition for weight loss" is recommended, and nutritional guidance that restricts calories is provided. On the other hand, in symptomatic Stage C and D heart failure, it is known that underweight patients who manifest poor nutrition, sarcopenia, and cardiac cachexia have a poor prognosis. This is referred to as the "Obesity paradox". In order to "avoid weight loss" in patients with heart failure, a paradigm shift to nutritional management to prevent weight loss is needed. Rather than prescribing uniform recommendation for salt reduction of 6 g/day or less, awareness of the behavior change stage model is attracting attention. In this setting, the value of salt restriction will need to be determined to determine the priority level of intervention for undernutrition versus the need to prevent congestive signs and symptoms. In the Intensive Care Unit (ICU)/Cardiac Care Unit (CCU) for acute heart failure, nutritional intervention should be considered within 48 h of admission. Key points are selection of access route, timing of intervention, and monitoring of side effects. In nutritional management at home and in end-of-life care, food is a reflection of an individual's values, as well as a source of joy and encouragement. The importance of digestive tract should also be recognized in heart failure from oral flail to intestinal edema, constipation, and the intestinal bacteria called the heart-gut axis. Finally, we would like to propose a new term "heart nutrition" for nutritional management in patients with heart failure in this review. Compared to the evidence for exercise therapy in heart failure, studies assessing nutritional management remain scarce and there is a need for research in this area in the future.
Collapse
Affiliation(s)
- Keisuke Kida
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan.
| | - Isao Miyajima
- Department of Clinical Nutrition, Chikamori Hospital, Kochi, Japan
| | - Norio Suzuki
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Barry H Greenberg
- Division of Cardiology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Yoshihiro J Akashi
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
48
|
Javan-Khoshkholgh A, Sassoon JC, Behbodikhah J, Dai W, Alemu S, Quadri S, Singh M, Savinova OV, Farajidavar A. Recording and analysis of slow waves of the small intestine of mice with heart failure. Neurogastroenterol Motil 2023; 35:e14514. [PMID: 36480434 DOI: 10.1111/nmo.14514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/18/2022] [Accepted: 11/23/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastrointestinal (GI) symptoms in heart failure (HF) patients are associated with increased morbidity and mortality. We hypothesized that HF reduces bioelectrical activity underlying peristalsis. In this study, we aimed to establish a method to capture and analyze slow waves (SW) in the small intestine in mice with HF. METHODS We established a model of HF secondary to coronary artery disease in mice overexpressing tissue-nonspecific alkaline phosphatase (TNAP) in endothelial cells. The myoelectric activity was recorded from the small intestine in live animals under anesthesia. The low- and high-frequency components of SW were isolated in MATLAB and compared between the control (n = 12) and eTNAP groups (n = 8). C-kit-positive interstitial cells of Cajal (ICC) and Pgp9.5-positive myenteric neurons were detected by immunofluorescence. Myenteric ganglia were assessed by hematoxylin and eosin (H&E) staining. RESULTS SW activity was successfully captured in vivo, with both high- and low-frequency components. Low-frequency component of SW was not different between endothelial TNAP (eTNAP) and control mice (mean[95% CI]: 0.032[0.025-0.039] vs. 0.040[0.028-0.052]). High-frequency component of SW showed a reduction eTNAP mice relative to controls (0.221[0.140-0.302] vs. 0.394[0.295-0.489], p < 0.01). Dysrhythmia was also apparent upon visual review of signals. The density of ICC and neuronal networks remained the same between the two groups. No significant reduction in the size of myenteric ganglia of eTNAP mice was observed. CONCLUSIONS A method to acquire SW activity from small intestines in vivo and isolate low- and high-frequency components was established. The results indicate that HF might be associated with reduced high-frequency SW activity.
Collapse
Affiliation(s)
- Amir Javan-Khoshkholgh
- Department of Materials Science and Biomedical Engineering, University of Wisconsin - Eau Claire, Eau Claire, Wisconsin, USA
| | - Joseph C Sassoon
- College of Engineering and Computing Sciences, Department of Electrical and Computer Engineering, New York Institute of Technology, Old Westbury, New York, USA
| | - Jennifer Behbodikhah
- College of Osteopathic Medicine, Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, New York, USA
| | - Wenchen Dai
- College of Engineering and Computing Sciences, Department of Electrical and Computer Engineering, New York Institute of Technology, Old Westbury, New York, USA
| | - Senayt Alemu
- College of Osteopathic Medicine, Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, New York, USA
| | - Saad Quadri
- College of Osteopathic Medicine, Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, New York, USA
| | - Mohnish Singh
- College of Osteopathic Medicine, Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, New York, USA
| | - Olga V Savinova
- College of Osteopathic Medicine, Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, New York, USA
| | - Aydin Farajidavar
- College of Engineering and Computing Sciences, Department of Electrical and Computer Engineering, New York Institute of Technology, Old Westbury, New York, USA
| |
Collapse
|
49
|
Ouyang L, Chen B, Liu X, Wang D, Li Y, Liao Y, Yeung KW, Liu X. Puerarin@Chitosan composite for infected bone repair through mimicking the bio-functions of antimicrobial peptides. Bioact Mater 2023; 21:520-530. [PMID: 36185735 PMCID: PMC9508162 DOI: 10.1016/j.bioactmat.2022.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
It is important to eliminate lipopolysaccharide (LPS) along with killing bacteria in periprosthetic joint infection (PJI) therapy for promoting bone repair due to its effect to regulate macrophages response. Although natural antimicrobial peptides (AMPs) offer a good solution, the unknown toxicity, high cost and exogenetic immune response hamper their applications in clinic. In this work, we fabricated a nanowire-like composite material, named P@C, by combining chitosan and puerarin via solid-phase reaction, which can finely mimic the bio-functions of AMPs. Chitosan, serving as the bacteria membrane puncture agent, and puerarin, serving as the LPS target agent, synergistically destroy the bacterial membrane structure and inhibit its recovery, thus endowing P@C with good antibacterial property. In addition, P@C possesses good osteoimmunomodulation due to its ability of LPS elimination and macrophage differentiation modulation. The in vivo results show that P@C can inhibit the LPS induced bone destruction in the Escherichia coli infected rat. P@C exhibits superior bone regeneration in Escherichia coli infected rat due to the comprehensive functions of its superior antibacterial property, and its ability of LPS elimination and immunomodulation. P@C can well mimic the functions of AMPs, which provides a novel and effective method for treating the PJI in clinic. P@C was fabricated through solid reaction with chitosan and puerarin. P@C punctures bacteria membrane and eliminates LPS, thus sterilizes bacteria. P@C improves bone formation of PEEK under infection via polarizing macrophage to M2.
Collapse
|
50
|
Pugliese NR, Pellicori P, Filidei F, De Biase N, Maffia P, Guzik TJ, Masi S, Taddei S, Cleland JGF. Inflammatory pathways in heart failure with preserved left ventricular ejection fraction: implications for future interventions. Cardiovasc Res 2023; 118:3536-3555. [PMID: 36004819 PMCID: PMC9897694 DOI: 10.1093/cvr/cvac133] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
Many patients with symptoms and signs of heart failure have a left ventricular ejection fraction ≥50%, termed heart failure with preserved ejection fraction (HFpEF). HFpEF is a heterogeneous syndrome mainly affecting older people who have many other cardiac and non-cardiac conditions that often cast doubt on the origin of symptoms, such as breathlessness, or signs, such as peripheral oedema, rendering them neither sensitive nor specific to the diagnosis of HFpEF. Currently, management of HFpEF is mainly directed at controlling symptoms and treating comorbid conditions such as hypertension, atrial fibrillation, anaemia, and coronary artery disease. HFpEF is also characterized by a persistent increase in inflammatory biomarkers. Inflammation may be a key driver of the development and progression of HFpEF and many of its associated comorbidities. Detailed characterization of specific inflammatory pathways may provide insights into the pathophysiology of HFpEF and guide its future management. There is growing interest in novel therapies specifically designed to target deregulated inflammation in many therapeutic areas, including cardiovascular disease. However, large-scale clinical trials investigating the effectiveness of anti-inflammatory treatments in HFpEF are still lacking. In this manuscript, we review the role of inflammation in HFpEF and the possible implications for future trials.
Collapse
Affiliation(s)
| | - Pierpaolo Pellicori
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| | - Francesco Filidei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples 80138, Italy
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Internal and Agricultural Medicine, Jagiellonian University, Collegium Medicum, Krakow 31-008, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - John G F Cleland
- Robertson Institute of Biostatistics and Clinical Trials Unit, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|