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
|
Palm CL, de Wit S, Gorter TM, Rienstra M, Vos MJ, Kema IP, van der Ley CP, Bakker SJL, Bakker BM, de Boer RA, van Veldhuisen DJ, Meijers WC, Westenbrink BD. Beyond the gut: Systemic levels of short-chain fatty acids are altered in patients with heart failure. Int J Cardiol 2025; 428:133124. [PMID: 40068788 DOI: 10.1016/j.ijcard.2025.133124] [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: 02/06/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND & AIM The gut microbiome produces short-chain fatty acids (SCFAs), which serve as a substantial energy source and provide a link between the microbiome and (cardiac) metabolism. It has been demonstrated that the composition of the microbiome is altered in patients with heart failure (HF), but whether circulating levels of SCFAs are altered in HF is unknown. METHODS & RESULTS Serum concentrations of the SCFAs acetate, propionate, and butyrate were measured in 205 patients with HF and in 54 healthy controls, using isotope dilution liquid chromatography-tandem mass spectrometry. Of the patients with HF, 99 had HF with a reduced ejection fraction (HFrEF) and 106 had HF with mildly-reduced or preserved ejection fraction (HFmrEF/HFpEF). Healthy controls were age and sex matched to the HFrEF patients. Serum concentrations of acetate and propionate were significantly lower in patients with HF than in healthy controls, whereas butyrate levels were higher in patients with HF. Analyses by HF type revealed that acetate and propionate levels were lower in both HFrEF and HFpEF/HFmrEF patients in comparison to healthy controls. However, butyrate levels were observed to be lower in patients with HFmrEF/HFpEF in comparison to healthy controls, while they were higher in patients with HFrEF. CONCLUSIONS In patients with HF, serum levels of acetate and propionate are lower across the HF spectrum, whereas serum butyrate levels are elevated in HFrEF, but lower in HFmrEF/HFpEF. These alterations in SCFA profiles suggest a microbiome-driven metabolic dysregulation, which appears to differ between HF subtypes.
Collapse
Affiliation(s)
- C L Palm
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Institute of Clinical Chemistry and Laboratory Medicine, Oldenburg Clinic, University of Oldenburg, Germany
| | - S de Wit
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - T M Gorter
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Rienstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M J Vos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - I P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - C P van der Ley
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - S J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B M Bakker
- Laboratory of Pediatrics, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - R A de Boer
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - D J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - W C Meijers
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - B D Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
3
|
Sayed A, Afify H, Munir M, ElGarhy I, Shazly O, ElRefaei M, Ahmed S, Amin AM, Amine OC, Elgendy IY. Prognostic value of lipid parameters among patients with heart failure: A systematic review and meta-analysis. ESC Heart Fail 2025. [PMID: 40411472 DOI: 10.1002/ehf2.15315] [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: 12/20/2024] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 05/26/2025] Open
Abstract
AIMS We sought to evaluate the prognostic value of different lipid parameters in patients with heart failure (HF). METHODS AND RESULTS Electronic databases including MEDLINE, Embase, CENTRAL, and Web of Science were searched to identify studies that reported the association of any of the four lipid parameters [total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and triglycerides] with mortality among patients with HF. A random-effects model was used to estimate the association per 10 mg/dL increment. The QUIPS tool was used to assess the risk of bias. Fifty-two studies enrolling 93 286 patients were included. On univariable analysis, higher levels of the four lipid parameters were associated with lower mortality: TC [hazard ratio/odds ratio (HR/OR): 0.94; 95% confidence interval (CI): 0.93 to 0.96], HDL-C (HR/OR: 0.89; 95% CI: 0.80 to 0.99), LDL-C (HR/OR: 0.93; 95% CI: 0.90 to 0.97) and triglycerides (HR/OR: 0.95; 95% CI: 0.92 to 0.99). On multivariable analysis, lower levels of TC (HR/OR: 0.95; 95% CI: 0.93 to 0.97) and LDL-C (HR/OR: 0.94; 95% CI: 0.89 to 0.99) were associated with lower mortality. CONCLUSIONS Higher levels of lipids parameters were associated with lower mortality in patients with HF. Lipid parameters may improve prognostication in predictive models for patients with HF. Because of the observational nature of included studies, no claims about the causal effect of changing lipid parameters can be made.
Collapse
Affiliation(s)
- Ahmed Sayed
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hesham Afify
- Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Malak Munir
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Omar Shazly
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Saeed Ahmed
- King Edward Medical University, Lahore, Pakistan
| | - Ahmed M Amin
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Islam Y Elgendy
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Sen I, Trzaskalski NA, Hsiao YT, Liu PP, Shimizu I, Derumeaux GA. Aging at the Crossroads of Organ Interactions: Implications for the Heart. Circ Res 2025; 136:1286-1305. [PMID: 40403108 DOI: 10.1161/circresaha.125.325637] [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: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/24/2025]
Abstract
Aging processes underlie common chronic cardiometabolic diseases such as heart failure and diabetes. Cross-organ/tissue interactions can accelerate aging through cellular senescence, tissue wasting, accelerated atherosclerosis, increased vascular stiffness, and reduction in blood flow, leading to organ remodeling and premature failure. This interorgan/tissue crosstalk can accelerate aging-related dysfunction through inflammation, senescence-associated secretome, and metabolic and mitochondrial changes resulting in increased oxidative stress, microvascular dysfunction, cellular reprogramming, and tissue fibrosis. This may also underscore the rising incidence and co-occurrence of multiorgan dysfunction in cardiometabolic aging in the population. Examples include interactions between the heart and the lungs, kidneys, liver, muscles, and brain, among others. However, this phenomenon can also present new translational opportunities for identifying diagnostic biomarkers to define early risks of multiorgan dysfunction, gain mechanistic insights, and help to design precision-directed therapeutic interventions. Indeed, this opens new opportunities for therapeutic development in targeting multiple organs simultaneously to disrupt the crosstalk-driven process of mutual disease acceleration. New therapeutic targets could provide synergistic benefits across multiple organ systems in the same at-risk patient. Ultimately, these approaches may together slow the aging process itself throughout the body. In the future, with patient-centered multisystem coordinated approaches, we can initiate a new paradigm of multiorgan early risk prediction and tailored intervention. With emerging tools including artificial intelligence-assisted risk profiling and novel preventive strategies (eg, RNA-based therapeutics), we may be able to mitigate multiorgan cardiometabolic dysfunction much earlier and, perhaps, even slow the aging process itself.
Collapse
Affiliation(s)
- Ilke Sen
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| | - Natasha A Trzaskalski
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Yung-Ting Hsiao
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
| | - Peter P Liu
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (I. Shimizu)
| | - Geneviève A Derumeaux
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| |
Collapse
|
5
|
Snelson M, Muralitharan RR, Liu CF, Markó L, Forslund SK, Marques FZ, Tang WHW. Gut-Heart Axis: The Role of Gut Microbiota and Metabolites in Heart Failure. Circ Res 2025; 136:1382-1406. [PMID: 40403109 PMCID: PMC12101525 DOI: 10.1161/circresaha.125.325516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is a global health issue with significant mortality and morbidity. There is increasing evidence that alterations in the gastrointestinal microbiome, gut epithelial permeability, and gastrointestinal disorders contribute to heart failure progression through various pathways, including systemic inflammation, metabolic dysregulation, and modulation of cardiac function. Moreover, several medications used to treat heart failure directly impact the microbiome. The relationship between the gastrointestinal tract and the heart is bidirectional, termed the gut-heart axis. It is increasingly understood that diet-derived microbial metabolites are key mechanistic drivers of the gut-heart axis. This includes, for example, trimethylamine N-oxide and short-chain fatty acids. This review discusses current insights into the interplay between heart failure, its associated risk factors, and the gut microbiome, focusing on key metabolic pathways, the role of dietary interventions, and the potential for gut-targeted therapies. Understanding these complex interactions could pave the way for novel strategies to mitigate heart failure progression and improve patient outcomes.
Collapse
Affiliation(s)
- Matthew Snelson
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Rikeish R. Muralitharan
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| | - Lajos Markó
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
| | - Sofia K. Forslund
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Francine Z. Marques
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - W. H. Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| |
Collapse
|
6
|
Yamashita T. The role of gut microbiota in cardiovascular diseases and their potential as novel therapeutic targets. J Cardiol 2025:S0914-5087(25)00126-1. [PMID: 40409712 DOI: 10.1016/j.jjcc.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/25/2025]
Abstract
Cardiovascular diseases (CVDs) including heart failure (HF) is a major health, medical and social issue that needs to be resolved in Japan's super-aged society. Recent clinical and basic studies suggest that the gut microbiota and their metabolites play critical roles in the onset and progression of CVDs. We explored changes in gut microbiota composition and metabolite levels among Japanese patients to investigate their association with CVDs. Changes in specific bacteria were observed, with a decrease in phylum Bacteroidetes and increases in order Lactobacillus or genus Streptococcus in coronary artery disease patients. For HF patients, a reduction in phylum Bacteroidetes and increases in phylum Actinobacteria (e.g. Bifidobacterium) and Proteobacteria (e.g. Escherichia, Shigella, and Klebsiella) were noted. Elevated levels of gut microbiota-associated metabolites, such as trimethylamine N-oxide (TMAO) and indoxyl sulfate, were observed in CVD patients, suggesting potential effects on organ functions. Many studies have linked higher plasma TMAO levels to worse prognoses in CVDs, including HF and renal failure. However, the clinical significance and therapeutic potential of these findings require further investigation. In this manuscript, the author aims to review the current status of research on gut microbiota in CVDs, with a primary focus on the microbes themselves and their related metabolites. Further research is essential to comprehensively understand these intricacies and establish clear cause-and-effect relationships, ultimately paving the way for the development of innovative therapies for CVDs.
Collapse
Affiliation(s)
- Tomoya Yamashita
- Division of Advanced Medical and Pharmaceutical Sciences, Graduate School of Science, Technology and Innovation, Kobe University, Japan.
| |
Collapse
|
7
|
Chen F, Guo Z, Chen Y, Li S, Chen P. Non-alcoholic fatty liver disease enhances the beneficial effect of renal denervation on gut microbiota aberrations in rats with heart failure. BMC Microbiol 2025; 25:311. [PMID: 40399816 PMCID: PMC12093722 DOI: 10.1186/s12866-025-04027-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Renal denervation (RDN) contributes to improving cardiac function by ameliorating aberrations of the gut microbiota, and non-alcoholic fatty liver disease (NAFLD) is associated with gut microbiota dysbiosis and is critically involved in the development of heart failure (HF). It is unclear whether the beneficial effect of RDN on gut microbiota in HF can be affected by NAFLD and whether this effect changes with the severity of NAFLD. METHODS HF Sprague Dawley rats induced by transverse aortic constriction were fed a high-fat-fructose diet and underwent RDN, and sequencing of 16S rRNA gene in fecal samples was detected. RESULTS The dissimilarity coefficients and sample distances of the intestinal microbiome were elevated in HF rats with NAFLD. After RDN, HF rats with NAFLD had fewer bacteria harmful to cardiac function, such as Alphaproteobacteria, Bacteroidota and Prevotella-9, and more bacteria beneficial to HF, such as Monoglobaceae, Proteobacteria and Monoglobales, than HF rats without NAFLD (all p < 0.05). This tendency also existed but was much less significant when compared between HF rats with non-alcoholic steatohepatitis (NASH) and without NAFLD. Predictive functional profiling of microbial communities revealed that after RDN, the abundance of membrane transport, environmental and genetic information processing was significantly higher, and glycan biosynthesis and metabolism was significantly lower in HF rats with NAFLD than in those without NAFLD. CONCLUSION NAFLD could further enhance the beneficial role of RDN in mitigating gut microbiota aberrations in HF rats by increasing beneficial bacteria and decreasing bacteria harmful to cardiac function, but this effect was not apparent in NASH rats.
Collapse
Affiliation(s)
- Fuyan Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, Guangdong, 510182, China
| | - Zhiqin Guo
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou, China
| | - Yufeng Chen
- Department of Cardiology, Maoming People's Hospital, Maoming, China
| | - Shun Li
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou, China
| | - Pingan Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, 1 Panfu Road, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
8
|
Deis T, Rossing K, Ersbøll M, Nielsen WH, Henriksen B, Hartmann B, Holst JJ, Kistorp C, Fudim M, Goetze JP, Jeppesen PB, Gustafsson F. Effect of a Meal on Invasive Hemodynamics and Plasma Incretin Levels in Patients With Heart Failure. Circ Heart Fail 2025; 18:e012630. [PMID: 40190265 DOI: 10.1161/circheartfailure.124.012630] [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: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND The importance of gastrointestinal hormones, including the incretins glucagon-like peptide-1 (GLP-1) and GIP (glucose-dependent insulinotropic polypeptide), in heart failure pathophysiology is debated. The postprandial incretin response and its relation to hemodynamic changes in patients with heart failure, however, remains unknown. METHODS A pulmonary artery catheter was placed in 14 patients with chronic heart failure and 10 healthy controls, who subsequently consumed a standardized meal (3.2 MJ). Hemodynamic measures and blood samples were collected over 120 minutes. Bowel wall thickness and superior mesenteric artery blood flow were measured by ultrasound. Gastrointestinal symptoms were assessed through questionnaires. RESULTS Patients, compared with controls, exhibited an impaired postprandial peak in cardiac index (2.8 versus 4.0 L/min per m², P<0.001) and a blunted cardiac index response in the postprandial period (baseline-substracted area under the curve, P=0.030). Patients had higher fasting total GLP-1 levels (14.0 versus 7.5 pmol/L, P=0.015) and a greater postprandial peak (33.5 versus 21.0 pmol/L, P=0.013). Fasting total GLP-1 concentrations correlated with central venous pressure (P=0.025). There was a trend toward a correlation between area under the curve total GLP-1 and cardiac index (P=0.054). There was no change in GIP concentrations. Patients had a higher gastrointestinal symptom burden (P=0.033), and an indigestion score that correlated with peak superior mesenteric artery blood flow (P=0.017). CONCLUSIONS Patients with heart failure showed a blunted cardiac index response to meal intake, elevated total GLP-1 levels that were associated with hemodynamic parameters, and increased gastrointestinal symptom burden.
Collapse
Affiliation(s)
- Tania Deis
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark (T.D., K.R., M.E., W.H.N., F.G.)
| | - Kasper Rossing
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark (T.D., K.R., M.E., W.H.N., F.G.)
| | - Mads Ersbøll
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark (T.D., K.R., M.E., W.H.N., F.G.)
| | - William Herrik Nielsen
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark (T.D., K.R., M.E., W.H.N., F.G.)
| | - Birthe Henriksen
- Department of Diagnostic Radiology, Rigshospitalet, Copenhagen, Denmark (B. Henriksen)
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B. Hartmann, J.J.H.)
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (B. Hartmann, J.J.H., J.P.G.)
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B. Hartmann, J.J.H.)
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (B. Hartmann, J.J.H., J.P.G.)
| | - Caroline Kistorp
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark (C.K.)
- Department of Clinical Medicine, University of Copenhagen, Denmark (F.G., C.K)
| | - Marat Fudim
- Duke University Medical Center, Durham, NC (M.F.)
- Duke Clinical Research Institute, Durham, NC (M.F.)
| | - Jens Peter Goetze
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark (B. Hartmann, J.J.H., J.P.G.)
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, University of Copenhagen, Denmark (J.P.G.)
| | - Palle Bekker Jeppesen
- Department of Medical Gastroenterology and Hepatology, Rigshospitalet, Copenhagen, Denmark (P.B.J.)
| | - Finn Gustafsson
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark (T.D., K.R., M.E., W.H.N., F.G.)
- Department of Clinical Medicine, University of Copenhagen, Denmark (F.G., C.K)
| |
Collapse
|
9
|
Appleby S, Purcell R. Circulating bacterial DNA in cardiovascular disease. Front Cardiovasc Med 2025; 12:1476165. [PMID: 40303615 PMCID: PMC12037610 DOI: 10.3389/fcvm.2025.1476165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Cardiovascular disease (CVD) remains a global health burden despite advances in prevention and treatment. Conventional biomarkers, while effective for a number of patient groups, fail to provide personalized diagnosis and prognosis, necessitating the exploration of novel markers. Advancements in sequencing technology have unveiled the role of cell-free DNA (cfDNA) as a reservoir of genetic information from all cells within the body, and associations between elevated cfDNA levels and CVD risk factors and status have been reported. Recent attention has turned to a subset of cfDNA, circulating bacterial DNA (cbDNA), derived from gut microbiota, as a potential biomarker. Investigations into microbial translocation from the gut, particularly the phenomenon of 'leaky gut,' reveal its association with CVD and provide a potential source for cbDNA. Here, we review the existing literature on cbDNA in CVD, highlighting its potential diagnostic and prognostic value. Current studies have largely been carried out in small, disparate cohorts, using different sample types and a range of methodologies. While cbDNA shows potential as a diagnostic and prognostic biomarker, the lack of consensus in methodologies and populations studied calls for standardized approaches and large cohorts to establish cbDNA as a reliable CVD biomarker. Future research should focus on identifying the source of cbDNA and its pathological relevance, utilizing advanced sequencing techniques and standardized cohorts for conclusive findings.
Collapse
Affiliation(s)
- Sarah Appleby
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Rachel Purcell
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
10
|
Tsai TY, Aldujeli A, Haq A, Murphy P, Unikas R, Sharif F, Garg S, Brilakis ES, Onuma Y, Serruys PW. Trimethylamine N-Oxide as a Biomarker for Left Ventricular Diastolic Dysfunction and Functional Remodeling After STEMI. Int J Mol Sci 2025; 26:3400. [PMID: 40244252 PMCID: PMC11989463 DOI: 10.3390/ijms26073400] [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: 03/05/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Despite successful primary percutaneous coronary intervention (PPCI), the incidence of heart failure (HF) following ST-elevation myocardial infarction (STEMI) remains high. We investigated using Trimethylamine N-oxide (TMAO), a gut microbiota-derived biomarker, to predict adverse functional left ventricular (LV) remodeling (FLVR) and/or diastolic dysfunction (DD), which are precursors of HF post-STEMI. This prospective, observational study enrolled 204 STEMI patients with multivessel coronary artery disease after PPCI. TMAO level was collected at the baseline and 3 months. An echocardiography was performed at the baseline and at 12 months. The primary endpoints were the number of patients developing Group 4 FLVR or ≥Grade II DD at 12 months. The median age was 65 [57.00, 76.00] and 39.7% were women. The primary endpoints occurred in 47 (23.0%) patients. Three months of TMAO can discriminate patients with/without ≥Grade II LV DD and FLVR Grade 4 with areas under the curve (AUC) of the ROC of 0.72 (95% CI: 0.63-0.81; p < 0.001) and 0.77 (95% CI: 0.63-0.91), respectively. Similar results were shown in the validation cohort of 31 patients. The addition of 3 months of TMAO to traditional risk factors significantly improved the AUCs from 0.675 to 0.736 for ≥Grade II DD and from 0.793 to 0.873 for FLVR Grade 4. In multivariable logistic regression, 3 months of TMAO was independently associated with ≥Grade II DD (OR: 1.29 (1.13-1.50), p < 0.001) and FLVR Grade 4 (OR: 1.28 (1.12-1.47), p < 0.001). Three months of TMAO is strongly associated with LV DD and adverse remodeling after STEMI and may help identifying such patients for early treatment.
Collapse
Affiliation(s)
- Tsung-Ying Tsai
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, H91 TK33 Galway, Ireland
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Ali Aldujeli
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (R.U.)
| | - Ayman Haq
- Abbott Northwestern Hospital/Minneapolis Heart Institute Foundation, Minneapolis, MN 55407, USA; (A.H.); (E.S.B.)
| | - Paddy Murphy
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, H91 TK33 Galway, Ireland
| | - Ramunas Unikas
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (R.U.)
| | - Faisal Sharif
- Department of Cardiology, University Hospital Galway, University of Galway, H91 YR71 Galway, Ireland
| | - Scot Garg
- Department of Cardiology, Royal Blackburn Hospital, Blackburn BB2 3HH, UK
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Emmanouil S. Brilakis
- Abbott Northwestern Hospital/Minneapolis Heart Institute Foundation, Minneapolis, MN 55407, USA; (A.H.); (E.S.B.)
| | - Yoshinobu Onuma
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, H91 TK33 Galway, Ireland
| | - Patrick W. Serruys
- CORRIB Research Centre for Advanced Imaging and Core Lab, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
11
|
Wang J, Yin J, Liu X, Liu Y, Jin X. Gut commensal bacterium Bacteroides vulgatus exacerbates helminth-induced cardiac fibrosis through succinate accumulation. PLoS Pathog 2025; 21:e1013069. [PMID: 40238740 PMCID: PMC12002503 DOI: 10.1371/journal.ppat.1013069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/21/2025] [Indexed: 04/18/2025] Open
Abstract
Trichinella spiralis (Ts) is known to cause cardiac fibrosis, which is a critical precursor to various heart diseases, and its progression is influenced by metabolic changes. However, the metabolic mechanisms remain unclear. Here, we observed that Ts-infected mice exhibited cardiac fibrosis along with elevated succinate levels in the heart using metabolomic analysis. Administration of succinate exacerbated fibrosis during Ts infection, while deficiency in succinate receptor 1 (Sucnr1) alleviated the condition, highlighting the role of the succinate-Sucnr1 axis in fibrosis development. Furthermore, metagenomics sequencing showed that Ts-infected mice had a higher abundance ratio of succinate-producing bacteria to succinate-consuming bacteria in the intestines. Notably, the succinate-producer Bacteroides vulgatus was enriched in Ts group. Oral supplementation with B. vulgatus aggravated Ts-induced cardiac fibrosis. In summary, our findings underscore the succinate-Sucnr1 axis as a critical pathway in helminth-induced cardiac fibrosis and highlight the potential of targeting this axis for therapeutic interventions. This study presents novel insights into the gut-heart axis, revealing innovative strategies for managing cardiovascular complications associated with helminth infections.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
- College of Animal Sciences, Jilin University, Changchun, China
| | - Jiali Yin
- The Second Hospital of Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yi Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Xuemin Jin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
12
|
Mocan D, Jipa R, Jipa DA, Lala RI, Rasinar FC, Groza I, Sabau R, Sulea Bratu D, Balta DF, Cioban ST, Puschita M. Unveiling the Systemic Impact of Congestion in Heart Failure: A Narrative Review of Multisystem Pathophysiology and Clinical Implications. J Cardiovasc Dev Dis 2025; 12:124. [PMID: 40278183 PMCID: PMC12028304 DOI: 10.3390/jcdd12040124] [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: 02/23/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Congestion is a key clinical feature of heart failure (HF), contributing to hospitalizations, disease progression, and poor outcomes. While traditionally considered a hemodynamic issue, congestion is now recognized as a systemic process affecting multiple organs. Renal dysfunction arises from impaired perfusion and sodium retention, leading to maladaptive left ventricular remodeling. Hepatic congestion contributes to cholestatic liver injury, while metabolic disturbances drive anemia, muscle wasting, and systemic inflammation. Additionally, congestion disrupts the intestinal barrier and immune function, exacerbating HF progression. Given its widespread impact, effective congestion management requires a shift from a cardiovascular-centered approach to a comprehensive, multidisciplinary strategy. Targeted decongestive therapy, metabolic and nutritional optimization, and immune modulation are crucial in mitigating congestion-related organ dysfunction. Early recognition and intervention are essential to slow disease progression, preserve functional capacity, and improve survival. Addressing HF congestion through personalized, evidence-based strategies is vital for optimizing long-term care and advancing treatment paradigms.
Collapse
Affiliation(s)
- Daniela Mocan
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.); (R.I.L.); (M.P.)
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 300310 Timisoara, Romania
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases of Timisoara, 300310 Timisoara, Romania
| | - Radu Jipa
- Faculty of Medicine, Department of “Life Sciences”, Vasile Goldis Western University of Arad, Romania 86, Liviu Rebreanu Street, 310048 Arad, Romania
- Arad County Clinical Emergency Hospital, 310037 Arad, Romania
| | - Daniel Alexandru Jipa
- Doctoral School, Victor Babes University of Timisoara, 300041 Timisoara, Romania;
- Victor Babes Clinical Hospital for Infectious Diseases and Pneumology of Timisoara, 300041 Timisoara, Romania
| | - Radu Ioan Lala
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.); (R.I.L.); (M.P.)
- Victor Babes Clinical Hospital for Infectious Diseases and Pneumology of Timisoara, 300041 Timisoara, Romania
| | - Florin Claudiu Rasinar
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 300310 Timisoara, Romania
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, Nr. 2, 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases of Timisoara, 300310 Timisoara, Romania
- Doctoral School, Victor Babes University of Timisoara, 300041 Timisoara, Romania;
| | - Iulia Groza
- Arad County Clinical Emergency Hospital, 310037 Arad, Romania
- Doctoral School, Victor Babes University of Timisoara, 300041 Timisoara, Romania;
| | - Ronela Sabau
- Arad County Clinical Emergency Hospital, 310037 Arad, Romania
| | | | - Diana Federica Balta
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.); (R.I.L.); (M.P.)
- Arad County Clinical Emergency Hospital, 310037 Arad, Romania
| | | | - Maria Puschita
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310025 Arad, Romania; (D.M.); (R.I.L.); (M.P.)
| |
Collapse
|
13
|
Huang L, Zhao X, Wang J, Guan J, Huang B, Feng J, Li X, Zhang Y, Zhang J. Gut microbiota and risk of heart failure in European population-A comprehensive Mendelian randomization study. ESC Heart Fail 2025. [PMID: 40098322 DOI: 10.1002/ehf2.15267] [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: 07/24/2023] [Revised: 06/01/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
AIMS Gut dysbiosis is proven to be involved in the pathogenesis and progression of heart failure (HF). Hindering the detrimental effects of gut-heart axis is an emerging trend. Our goal is to investigate the causal relationship between gut microbiota and HF, with the aim of facilitating future exploration of microbiome-targeted approaches to prevent and delay the progression of HF. METHODS AND RESULTS Two-sample Mendelian randomization (MR) analysis was applied to investigate the causal association of the gut microbiome with HF among individuals of European ancestry. Genetic variants associated with the 196 bacterial taxa from MiBioGen consortium were used as exposure data, summary statistics for HF derived from Heart Failure Molecular Epidemiology for Therapeutic Targets (HERMES) consortium were used as outcome data. Five MR methods were applied, including inverse variance weighted, maximum likelihood, MR-Egger, weighted median, and weighted mode. Reverse causality of instrumental variables (IVs) was tested by MR Steiger test of directionality. Strength of IVs was evaluated by F-statistics. Cochrane's Q test, MR-Egger regression analysis, and MR Pleiotropy RESidual Sum and Outlier (MR-PRESSO) tests were used to detect heterogeneity and pleiotropy. Leave-one-out method was used for testing the stability of results. Seven microbiomes were found to be associated with HF. Five of them were associated with higher risks of developing HF, these included Order_Selenomonadales (odds ratio [OR] = 1.11, P = 0.024), Family_Peptococcaceae (OR = 1.07, P = 0.045), Genus_Eubacterium eligens group (OR = 1.14, P = 0.022), Genus_Eubacterium oxidoreducens group (OR = 1.12, P = 0.011) and Genus_Flavonifractor (OR = 1.14, P = 0.012). Genus_Anaerostipes and Order_Bacillales were associated with lower risks of HF (OR = 0.90, P = 0.014; OR = 0.95, P = 0.042, respectively). Evidence of pleiotropy or heterogeneity was not observed. CONCLUSIONS We identified seven intestinal microbiomes that were causally associated with HF at the level of gene prediction. This study will help with the discovery of potential preventive and therapeutic targets for HF.
Collapse
Affiliation(s)
- Liyan Huang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xuemei Zhao
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jing Wang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jingyuan Guan
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Boping Huang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiayu Feng
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xinqing Li
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yuhui Zhang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jian Zhang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
- Key Laboratory of Clinical Research for Cardiovascular Medications, National Health Committee, Beijing, China
| |
Collapse
|
14
|
Ranjan P, Goswami SK, Dutta RK, Colin K, Pal HC, Zhang Q, Lal H, Prasad R, Verma SK. Hypertrophic heart failure promotes gut dysbiosis and gut leakage in interleukin 10-deficient mice. Am J Physiol Heart Circ Physiol 2025; 328:H447-H459. [PMID: 39854049 DOI: 10.1152/ajpheart.00323.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/10/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Heart failure (HF) is a leading cause of death worldwide. We have shown that pressure overload (PO)-induced inflammatory cell recruitment leads to heart failure in IL-10 knockout (KO) mice. However, it is unclear whether PO-induced inflammatory cells also target the gut mucosa, causing gut dysbiosis and leakage. We hypothesized that transverse aortic constriction (TAC) exacerbates immune cell homing to the gut (small intestine and colon), promoting dysbiosis and gut leakage in IL-10 KO mice. HF was induced in 8- to 10-wk-old C57BL/6J wild-type (WT) and B6.129P2-Il10tm1Cgn/J mutant (IL-10 KO) male and female mice by TAC and cardiac function was measured using visual sonics VEVO 3100. Fourteen days post-TAC, levels of monocytes, macrophages, neutrophils, and proinflammatory cytokines were measured in blood and gut. Gut dysbiosis was assessed via 16S rRNA sequencing in feces at 56 days post-TAC. IL-10 KO mice showed worsened cardiac dysfunction post-TAC. TAC worsened monocytes, and neutrophils infiltration in systemic circulation and facilitated their homing to the gut in IL-10 KO mice. Intriguingly, proinflammatory cytokines level was increased in blood, and gut of IL-10 KO mice following TAC. Furthermore, IL-10 expression was reduced in the colon of WT mice post-TAC. Moreover, TAC exacerbated gut dysbiosis in IL-10 KO mice. Finally, an impaired intestinal permeability was noted in IL-10 KO mice post-TAC. In conclusion, TAC-induced systemic inflammation leads to gut dysbiosis and impaired gut permeability in IL-10 KO mice, indicating IL-10's potential role in regulating intestinal integrity and microbiota balance during heart failure.NEW & NOTEWORTHY IL-10, crucial for systemic inflammation regulation and gut mucosal homeostasis, was investigated using IL-10 knockout (KO) mice. Exacerbated gut inflammation was observed post-transverse aortic constriction (TAC) in IL-10-depleted mice, whereas wild-type (WT) mice showed reduced IL-10 gene expression in colon and ileum. TAC induced gut dysbiosis and leakage in IL-10 KO mice, suggesting a link between enhanced inflammatory signaling in heart failure and multi-organ damage via gut dysbiosis and leakage.
Collapse
Affiliation(s)
- Prabhat Ranjan
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Sumanta Kumar Goswami
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Roshan Kumar Dutta
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Karen Colin
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Harish Chandra Pal
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Qinkun Zhang
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
15
|
Davison S, Mascellani Bergo A, Ward Z, Sackett A, Strykova A, Jaimes JD, Travis D, Clayton JB, Murphy HW, Danforth MD, Smith BK, Blekhman R, Fuh T, Niatou Singa FS, Havlik J, Petrzelkova K, Gomez A. Cardiometabolic disease risk in gorillas is associated with altered gut microbial metabolism. NPJ Biofilms Microbiomes 2025; 11:33. [PMID: 39984469 PMCID: PMC11845621 DOI: 10.1038/s41522-025-00664-3] [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: 10/24/2023] [Accepted: 02/03/2025] [Indexed: 02/23/2025] Open
Abstract
Cardiometabolic disease is the leading cause of death in zoo apes; yet its etiology remains unknown. Here, we investigated compositional and functional microbial markers in fecal samples from 57 gorillas across U.S. zoos, 20 of which are diagnosed with cardiovascular disease, in contrast with 17 individuals from European zoos and 19 wild gorillas from Central Africa. Results show that zoo-housed gorillas in the U.S. exhibit the most diverse gut microbiomes and markers of increased protein and carbohydrate fermentation, at the expense of microbial metabolic traits associated with plant cell-wall degradation. Machine learning models identified unique microbial traits in U.S. gorillas with cardiometabolic distress; including reduced metabolism of sulfur-containing amino acids and hexoses, increased abundance of potential enteric pathogens, and low fecal butyrate and propionate production. These findings show that cardiometabolic disease in gorillas is potentially associated with altered gut microbial function, influenced by zoo-specific diets and environments.
Collapse
Affiliation(s)
- Samuel Davison
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Anna Mascellani Bergo
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Zoe Ward
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA
| | - April Sackett
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA
| | - Anna Strykova
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - José Diógenes Jaimes
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Dominic Travis
- The Marine Mammal Center, Sausalito, CA, USA
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jonathan B Clayton
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Hayley W Murphy
- HWM and MDD: Great Ape Heart Project, Detroit Zoological Society, Royal Oak, MI, USA
| | - Marietta D Danforth
- HWM and MDD: Great Ape Heart Project, Detroit Zoological Society, Royal Oak, MI, USA
| | | | - Ran Blekhman
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Terence Fuh
- WWF Central African Republic, Bayanga, Central African Republic
| | | | - Jaroslav Havlik
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic.
| | - Klara Petrzelkova
- Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic.
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Brno, Czech Republic.
- Liberec Zoo, Liberec, Czech Republic.
| | - Andres Gomez
- Department of Animal Science, University of Minnesota, Saint Paul, MN, USA.
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
16
|
Shen S, Tian B, Zhang H, Wang YC, Li T, Cao Y. Heart Failure and Gut Microbiota: What Is Cause and Effect? RESEARCH (WASHINGTON, D.C.) 2025; 8:0610. [PMID: 39981296 PMCID: PMC11839986 DOI: 10.34133/research.0610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Emerging evidence highlights the central role of gut microbiota in maintaining physiological homeostasis within the host. Disruptions in gut microbiota can destabilize systemic metabolism and inflammation, driving the onset and progression of cardiometabolic diseases. In heart failure (HF), intestinal dysfunction may induce the release of endotoxins and metabolites, leading to dysbiosis and exacerbating HF through the gut-heart axis. Understanding the relationship between gut microbiota and HF offers critical insights into disease mechanisms and therapeutic opportunities. Current research highlights promising potential to improve patient outcomes by restoring microbiota balance. In this review, we summarize the current studies in understanding the gut microbiota-HF connection and discuss avenues for future investigation.
Collapse
Affiliation(s)
- Shichun Shen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Beiduo Tian
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Haizhu Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yu-Chen Wang
- Department of Medicine, Division of Cardiology, Department of Microbiology, Immunology and Molecular Genetics, and Department of Human Genetics,
University of California, Los Angeles, CA, USA
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics,
West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yang Cao
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
17
|
Luk A, Teijeiro-Paradis R, Kochan A, Billia F, Douflé G, Magder S, Mendelson AA, McGuinty C, Granton J. The Etiology and Management of Critical Acute Right Heart Failure. Can J Cardiol 2025:S0828-282X(25)00113-8. [PMID: 39938716 DOI: 10.1016/j.cjca.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
Right ventricular failure contributes to the morbidity and mortality of acute myocardial function, massive pulmonary embolism, and chronic pulmonary hypertension. Understanding how the normal physiology of the right ventricle (RV) is disrupted is integral to managing patients who present with RV decompensation. Therapeutic advances in mechanical circulatory support, pharmacotherapies to reduce afterload, mechanical and chemical lytic therapies for acute pulmonary embolism have improved outcomes of patients by offloading the RV. In this report we provide an overview of the physiology of the RV, medical management (volume optimization, hemodynamic targets, rhythm management), along with critical care-specific topics (induction with mechanical ventilation, sedation strategies, and mechanical circulatory support) and provide a framework for managing patients who present with leveraging principles of preload, contractility, and afterload. Last, because of the complexity of right ventricular failure management, and the complexity of presentation, we also discuss the role of team-based approach (cardiogenic shock and pulmonary embolism response teams), and highlight its benefits at improving outcomes.
Collapse
Affiliation(s)
- Adriana Luk
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada.
| | | | - Andrew Kochan
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Filio Billia
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Ghislaine Douflé
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| | - Sheldon Magder
- Department of Critical Care, McGill University Health Centre, Montreal, Quebec, Canada
| | - Asher A Mendelson
- Section of Critical Care, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - John Granton
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Shi Z, Yun M, Liu H, Li S, Zhang X. Impact of geriatric nutritional risk index and diabetes mellitus on prognosis in ischaemic heart failure with reduced ejection fraction. Sci Rep 2025; 15:4226. [PMID: 39905242 PMCID: PMC11794575 DOI: 10.1038/s41598-025-88950-0] [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: 10/04/2024] [Accepted: 02/01/2025] [Indexed: 02/06/2025] Open
Abstract
It is unclear whether diabetes mellitus (DM) affects the role of malnutrition in heart failure (HF). We evaluated the effect of the geriatric nutritional risk index (GNRI) on HF prognosis and DM's role in this relationship. This single-centre retrospective cohort study included 540 HF patients with nutritional data grouped by DM status and GNRI score. The primary endpoint was all-cause mortality. Eighty-four patients (15.6%) were classified as malnourished (GNRI ≤ 98). Over a median follow-up of 4.0 years, 102 patients died. The DM/low GNRI (L-GNRI) group had the highest risk of all-cause death (HRadj: 3.253, 95% CI 1.643-6.474, P < 0.001) and cardiac death (HRadj: 3.411, 95% CI 1.606-7.243, P < 0.001) compared to the non-DM/high GNRI group. The adverse impact of L-GNRI was more pronounced in DM than in non-DM patients (Pinteraction < 0.05). In the total population and DM subgroup, GNRI was independently associated with an increased risk of all-cause and cardiac death after adjustment (all P < 0.05). In patients with DM, the GNRI classification significantly enhanced the predictive value of the model (all P < 0.05). A negative correlation between GNRI and HbA1c was observed only in patients with DM. Patients with HF with DM and malnutrition had the poorest prognosis. Poor glycemic control is related to increased malnutrition risk.
Collapse
Affiliation(s)
- Zhiyong Shi
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingkai Yun
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoli Zhang
- Department of Nuclear Medicine, Molecular Imaging Lab, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
19
|
Carretero Gómez J, Galeano Fernández TF, Vidal Ríos AS, Tolosa Álvarez S, Sánchez Herrera M, García Carrasco C, Monreal Periañez FJ, González González P, Arévalo Lorido JC. Malnutrition in heart failure. The importance of assessing for congestion and sarcopenia. Rev Clin Esp 2025; 225:92-100. [PMID: 39615877 DOI: 10.1016/j.rceng.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/29/2024] [Indexed: 12/10/2024]
Abstract
AIM This work aims to describe nutrition and sarcopenia in inpatients with heart failure (HF). It also aims to assess factors associated with in-hospital and short-term prognosis related to nutrition and sarcopenia. METHODS We collected data on consecutive HF patients admitted to a single center's internal medicine ward. Patients were recruited in May and October 2021. Malnutrition was determined by the Mini Nutritional Assessment-Short Form (MNA-SF) and sarcopenia by the screening test, SARC-F scale, and handgrip strength test. RESULTS 190 patients were analyzed, mean age 82.1 (±8.2), 54.2% women, median follow up 106 days. Patients were classified into three groups based on MNA-SF score: group 1 (12-14 points, no risk) included 50 patients, group 2 (8-12 points, high risk of malnutrition) included 81 patients, group 3 (0-7 points, malnourished) included 59 patients. Group 3 had significantly more inflammation (lower albumin and higher C-reactive Protein (CRP)) and congestion (measured by NT-ProBNP levels). In-hospital mortality was related to poor muscle function, CRP, and NT-ProBNP, but not to malnutrition. The poorest short-term outcomes were related to malnutrition and comorbidity. However, when the variable of muscle function was introduced, it could act as a poor prognostic factor related to CRP and NT-ProBNP, which were the main determinants of survival. CONCLUSION In malnourished patients with HF, inflammation and congestion were related to in-hospital mortality. Malnutrition along with comorbidity may play a role in decreasing short-term survival. Early identification through proactive nutritional and functional screenings should be a key element of assessing HF patients.
Collapse
Affiliation(s)
- J Carretero Gómez
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain.
| | - T F Galeano Fernández
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | - A S Vidal Ríos
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | - S Tolosa Álvarez
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | - M Sánchez Herrera
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | - C García Carrasco
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | - F J Monreal Periañez
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | - P González González
- Internal Medicine Department. University Hospital Complex of Badajoz, Badajoz, Spain
| | | |
Collapse
|
20
|
Escalante J, Artaiz O, Diwakarla S, McQuade RM. Leaky gut in systemic inflammation: exploring the link between gastrointestinal disorders and age-related diseases. GeroScience 2025; 47:1-22. [PMID: 39638978 PMCID: PMC11872833 DOI: 10.1007/s11357-024-01451-2] [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: 08/25/2023] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
Global average life expectancy has steadily increased over the last several decades and is projected to reach ~ 77 years by 2050. As it stands, the number of people > 60 years currently outnumbers children younger than 5 years, and by 2050, it is anticipated that the global population of people aged > 60 years will double, surpassing 2.1 billion. This demographic shift in our population is expected to have substantial consequences on health services globally due to the disease burden associated with aging. Osteoarthritis, chronic obstructive pulmonary disease, diabetes, cardiovascular disease, and cognitive decline associated with dementia are among the most common age-related diseases and contribute significantly to morbidity and mortality in the aged population. Many of these age-related diseases have been linked to chronic low-grade systemic inflammation which often accompanies aging. Gastrointestinal barrier dysfunction, also known as "leaky gut," has been shown to contribute to systemic inflammation in several diseases including inflammatory bowel disease and irritable bowel syndrome, but its role in the development and/or progression of chronic low-grade systemic inflammation during aging is unclear. This review outlines current literature on the leaky gut in aging, how leaky gut might contribute to systemic inflammation, and the links between gastrointestinal inflammatory diseases and common age-related diseases to provide insight into a potential relationship between the intestinal barrier and inflammation.
Collapse
Affiliation(s)
- Jonathan Escalante
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia
| | - Olivia Artaiz
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia
| | - Shanti Diwakarla
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia
| | - Rachel M McQuade
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia.
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), The Melbourne University and Western Health, Melbourne, VIC, 3021, Australia.
| |
Collapse
|
21
|
Lombardo SF, Ferasin H, Ferasin L. Subcutaneous Furosemide Therapy for Chronic Management of Refractory Congestive Heart Failure in Dogs and Cats. Animals (Basel) 2025; 15:358. [PMID: 39943128 PMCID: PMC11815753 DOI: 10.3390/ani15030358] [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: 01/07/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Reduced efficacy of oral diuretics in the treatment of congestive heart failure (CHF) can be secondary to reduced enteral drug absorption. The aim of this study was to determine the efficacy of subcutaneous (SC) furosemide administration to control the signs of refractory CHF in dogs and cats and to assess the feasibility of this route of administration. The clinical records of 13 dogs and 17 cats with a history of refractory CHF treated with subcutaneous (SC) furosemide were reviewed retrospectively. Administration of SC furosemide was offered as an alternative therapy when animals experienced an unsatisfactory clinical response to oral diuretics despite multiple dose adjustments and when, for this reason, pet owners were considering euthanasia. The satisfactory control of the animal's breathing rate and effort and overall pet owner's satisfaction were observed in all cases. Following administration of SC furosemide, the median survival time was 106 (95% CI: 22-154) days in dogs and 89 (95% CI: 35 to 749) days in cats. This study showed that furosemide administered subcutaneously appears to be an efficacious and feasible therapeutic option for providing control of the signs of cardiac congestion in both dogs and cats with a previous unsatisfactory response to oral diuresis.
Collapse
Affiliation(s)
| | - Heidi Ferasin
- Specialist Veterinary Cardiology Consultancy Ltd., Four Marks GU34 5AA, UK;
| | - Luca Ferasin
- The Ralph Veterinary Referral Centre, Marlow SL7 1YG, UK;
- Specialist Veterinary Cardiology Consultancy Ltd., Four Marks GU34 5AA, UK;
| |
Collapse
|
22
|
Abdulrahim AO, Doddapaneni NSP, Salman N, Giridharan A, Thomas J, Sharma K, Abboud E, Rochill K, Shreelakshmi B, Gupta V, Lakkimsetti M, Mowo-Wale A, Ali N. The gut-heart axis: a review of gut microbiota, dysbiosis, and cardiovascular disease development. Ann Med Surg (Lond) 2025; 87:177-191. [PMID: 40109640 PMCID: PMC11918638 DOI: 10.1097/ms9.0000000000002789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/20/2024] [Indexed: 03/22/2025] Open
Abstract
Background Cardiovascular diseases (CVDs) are a major cause of morbidity and mortality worldwide and there are strong links existing between gut health and cardiovascular health. Gut microbial diversity determines gut health. Dysbiosis, described as altered gut microbiota, causes bacterial translocations and abnormal gut byproducts resulting in systemic inflammation. Objective To review the current literature on the relationships between gut microbiota, dysbiosis, and CVD development, and explore therapeutic methods to prevent dysbiosis and support cardiovascular health. Summary Dysbiosis increases levels of pro-inflammatory substances while reducing those of anti-inflammatory substances. This accumulative inflammatory effect negatively modulates the immune system and promotes vascular dysfunction and atherosclerosis. High Firmicutes to Bacteroidetes ratios, high trimethylamine-n-oxide to short-chain fatty acid ratios, high indole sulfate levels, low cardiac output, and polypharmacy are all associated with worse cardiovascular outcomes. Supplementation with prebiotics and probiotics potentially alleviates some CVD risk. Blood and stool samples may be used in clinical practice to quantify and qualify gut bacterial ratios and byproducts, assess patients' risk for adverse cardiovascular outcomes, and track their gut health progress. Further research is required to set population-based cutoffs for normal and abnormal gut microbiota and byproduct ratios.
Collapse
Affiliation(s)
| | | | - Nadhra Salman
- Department of Internal Medicine, Baqai Medical University, Karachi, Pakistan
| | | | | | - Kavya Sharma
- Maharishi Markandeshwar Medical College and Hospital, Himachal Pradesh, India
| | - Elias Abboud
- Faculty of Medicine, University of Saint Joseph, Beirut, Lebanon
| | | | - B Shreelakshmi
- Navodaya Medical College Hospital & Research Centre, Karnataka, India
| | | | | | | | - Noor Ali
- Dubai Medical College, Dubai, United Arab Emirates
| |
Collapse
|
23
|
Rosas PC, Neves LAA, Patel N, Tran D, Pereira CH, Bonilla KR, Zheng J, Sun J, Alvarado FJ, Banach K. Early pathological mechanisms in a mouse model of heart failure with preserved ejection fraction. Am J Physiol Heart Circ Physiol 2024; 327:H1524-H1543. [PMID: 39485297 PMCID: PMC11684889 DOI: 10.1152/ajpheart.00318.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) constitutes more than half of all HF cases, yet evidence-based therapies remain lacking due to limited understanding of its underlying pathological mechanisms. Our study aimed to uncover early pathological mechanisms in HFpEF by exposing mice to dietary conditions resembling a Western diet-rich in fats, salt, and low in fiber-alongside excess mineralocorticoids to replicate significant aspects of human HFpEF. Echocardiography was performed at both 3-wk and 6-wk intervals postchallenge, revealing cardiac alterations as early as 3 wk. While ejection fraction remained preserved, mice exhibited signs of diastolic dysfunction, reduced stroke volume, and left atrial enlargement. In addition, changes in pulmonary flow velocities were noted by the 3-wk mark, suggesting elevated pulmonary pressure. Extracardiac comorbidities included organ congestion, increased adiposity, impaired glucose tolerance, and hypercholesterolemia. Molecular analyses unveiled evidence of low-grade inflammation, oxidative stress, and impaired NO-cGMP-PKG signaling, contributing to the observed decrease in titin phosphorylation, thereby impacting myocardial stiffness. In addition, impaired nitric oxide (NO) signaling might have influenced the alterations observed in coronary flow reserve. Moreover, dysregulation of calcium signaling in cardiomyocytes and reduced sarcoplasmic reticulum (SR) load were observed. Interestingly, elevated phosphorylation of cMyBP-C was linked to preserved ejection fraction despite reduced SR load. We also observed intestinal atrophy, possibly due to a high-fat diet, low dietary fiber intake, and diminished gut perfusion, potentially contributing to systemic low-grade inflammation. These findings reveal how excess mineralocorticoid salt-induced hypertension and dietary factors, like high-fat and low-fiber intake, contribute to cardiac dysfunction and metabolic disturbances, offering insights into early HFpEF pathology in this model.NEW & NOTEWORTHY Our study demonstrates that feeding mice a Western diet rich in fat and salt and low in fiber alongside excess mineralocorticoids replicates aspects of human HFpEF. Cardiac alterations including diastolic dysfunction and decreased stroke volume with preserved ejection fraction were observed. Extracardiac effects included organ congestion, adiposity, glucose intolerance, and intestinal atrophy. Molecular analysis revealed inflammation, oxidative stress, impaired NO-cGMP-PKG signaling pathways, and altered calcium signaling in cardiomyocytes, shedding light on early pathological changes in HFpEF.
Collapse
Grants
- HL155762 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL155762 NHLBI NIH HHS
- R01 HL132871 NHLBI NIH HHS
- R01 HL164453 NHLBI NIH HHS
- S10 OD027016 NIH HHS
- HL155241-02S1 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL161070 NHLBI NIH HHS
- HL164453 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL167195 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K01 HL155241 NHLBI NIH HHS
- HL161070 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL155241 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- CDA849387 American Heart Association (AHA)
- R01 HL167195 NHLBI NIH HHS
- University of Illinois
Collapse
Affiliation(s)
- Paola C Rosas
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Liomar A A Neves
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States
- AbbVie Inc, Chicago, Illinois, United States
| | - Nisha Patel
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Duyen Tran
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Carlos H Pereira
- Department of Internal Medicine/Cardiology, Rush University Medical Center, Chicago, Illinois, United States
| | - Karina R Bonilla
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Jingjing Zheng
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- UIC Cancer Center, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Francisco J Alvarado
- Department of Medicine and Cardiovascular Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Kathrin Banach
- Department of Internal Medicine/Cardiology, Rush University Medical Center, Chicago, Illinois, United States
| |
Collapse
|
24
|
Sun N, Ogulur I, Mitamura Y, Yazici D, Pat Y, Bu X, Li M, Zhu X, Babayev H, Ardicli S, Ardicli O, D'Avino P, Kiykim A, Sokolowska M, van de Veen W, Weidmann L, Akdis D, Ozdemir BG, Brüggen MC, Biedermann L, Straumann A, Kreienbühl A, Guttman-Yassky E, Santos AF, Del Giacco S, Traidl-Hoffmann C, Jackson DJ, Wang DY, Lauerma A, Breiteneder H, Zhang L, O'Mahony L, Pfaar O, O'Hehir R, Eiwegger T, Fokkens WJ, Cabanillas B, Ozdemir C, Kistler W, Bayik M, Nadeau KC, Torres MJ, Akdis M, Jutel M, Agache I, Akdis CA. The epithelial barrier theory and its associated diseases. Allergy 2024; 79:3192-3237. [PMID: 39370939 DOI: 10.1111/all.16318] [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: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, over 350,000 new chemical substances have been introduced to the lives of humans. In recent years, the epithelial barrier theory came to light explaining the growing prevalence and exacerbations of these diseases worldwide. It attributes their onset to a functionally impaired epithelial barrier triggered by the toxicity of the exposed substances, associated with microbial dysbiosis, immune system activation, and inflammation. Diseases encompassed by the epithelial barrier theory share common features such as an increased prevalence after the 1960s or 2000s that cannot (solely) be accounted for by the emergence of improved diagnostic methods. Other common traits include epithelial barrier defects, microbial dysbiosis with loss of commensals and colonization of opportunistic pathogens, and circulating inflammatory cells and cytokines. In addition, practically unrelated diseases that fulfill these criteria have started to emerge as multimorbidities during the last decades. Here, we provide a comprehensive overview of diseases encompassed by the epithelial barrier theory and discuss evidence and similarities for their epidemiology, genetic susceptibility, epithelial barrier dysfunction, microbial dysbiosis, and tissue inflammation.
Collapse
Affiliation(s)
- Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xiangting Bu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xueyi Zhu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lukas Weidmann
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deniz Akdis
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Kreienbühl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St. Thomas' Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - David J Jackson
- Guy's Severe Asthma Centre, Guy's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore City, Singapore
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Robyn O'Hehir
- Allergy, Asthma & Clinical Immunology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Wytske J Fokkens
- Department of Otorhinolaryngology & Head and Neck Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Biosanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | - Walter Kistler
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Medical Committee International Ice Hockey Federation (IIHF), Zurich, Switzerland
| | - Mahmut Bayik
- Department of Internal Medicine and Hematology, Marmara University, Istanbul, Turkey
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Maria J Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
25
|
Zhong L, Yin H, Tang Z, Xiao Z. Evaluating the prognostic significance of RV-PA coupling in mitral valve repair via edge-to-edge technique: A systematic review and meta-analysis. Asian J Surg 2024:S1015-9584(24)02781-7. [PMID: 39613638 DOI: 10.1016/j.asjsur.2024.11.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024] Open
Affiliation(s)
- Lin Zhong
- Department of Cardio-Thoracic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, 610017, China; Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| | - Hesong Yin
- Department of Cardio-Thoracic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, 610017, China; Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| | - Zichun Tang
- Department of Cardio-Thoracic Surgery, North Sichuan Medical College, Nanchong, Sichuan, 637000, China.
| | - Zongwei Xiao
- Department of Cardio-Thoracic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, 610017, China; Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| |
Collapse
|
26
|
Matacchione G, Piacenza F, Pimpini L, Rosati Y, Marcozzi S. The role of the gut microbiota in the onset and progression of heart failure: insights into epigenetic mechanisms and aging. Clin Epigenetics 2024; 16:175. [PMID: 39614396 PMCID: PMC11607950 DOI: 10.1186/s13148-024-01786-9] [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: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The gut microbiota (GM) plays a critical role in regulating human physiology, with dysbiosis linked to various diseases, including heart failure (HF). HF is a complex syndrome with a significant global health impact, as its incidence doubles with each decade of life, and its prevalence peaks in individuals over 80 years. A bidirectional interaction exists between GM and HF, where alterations in gut health can worsen the disease's progression. MAIN BODY The "gut hypothesis of HF" suggests that HF-induced changes, such as reduced intestinal perfusion and altered gut motility, negatively impact GM composition, leading to increased intestinal permeability, the release of GM-derived metabolites into the bloodstream, and systemic inflammation. This process creates a vicious cycle that further deteriorates heart function. GM-derived metabolites, including trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and secondary bile acids (BAs), can influence gene expression through epigenetic mechanisms, such as DNA methylation and histone modifications. These epigenetic changes may play a crucial role in mediating the effects of dysbiotic gut microbial metabolites, linking them to altered cardiac health and contributing to the progression of HF. This process is particularly relevant in older individuals, as the aging process itself has been associated with both dysbiosis and cumulative epigenetic alterations, intensifying the interplay between GM, epigenetic changes, and HF, and further increasing the risk of HF in the elderly. CONCLUSION Despite the growing body of evidence, the complex interplay between GM, epigenetic modifications, and HF remains poorly understood. The dynamic nature of epigenetics and GM, shaped by various factors such as age, diet, and lifestyle, presents significant challenges in elucidating the precise mechanisms underlying this complex relationship. Future research should prioritize innovative approaches to overcome these limitations. By identifying specific metabolite-induced epigenetic modifications and modulating the composition and function of GM, novel and personalized therapeutic strategies for the prevention and/or treatment of HF can be developed. Moreover, targeted research focusing specifically on older individuals is crucial for understanding the intricate connections between GM, epigenetics, and HF during aging.
Collapse
Affiliation(s)
- Giulia Matacchione
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60127, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121, Ancona, Italy
| | | | - Yuri Rosati
- Pneumologia, IRCCS INRCA, 60027, Osimo, Italy
| | - Serena Marcozzi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, 60121, Ancona, Italy.
| |
Collapse
|
27
|
Hao R, Zheng Y, Zhao Q, Chen J, Fan R, Chen P, Yin N, Qin H. Evaluation value of ultrasound on gastrointestinal function in patients with acute heart failure. Front Cardiovasc Med 2024; 11:1475920. [PMID: 39654944 PMCID: PMC11625756 DOI: 10.3389/fcvm.2024.1475920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Objective To study the changes in gastrointestinal wall thickness, blood flow, motility, and symptoms in patients with acute heart failure, and to assess gastrointestinal function by ultrasound. Methods In this study, patients diagnosed with acute heart failure were selected as the study group, and healthy individuals were selected as the control group. Both groups collected general data and completed the Chinese version of the gastrointestinal symptom rating scale. Ultrasonography was used to measure several abdominal vascular and gastrointestinal-related indicators. Statistical analysis used grouped comparison and correlation analysis. Results The study group scored higher than the control group in total score, lower abdominal symptom score, constipation score, and difficult defecation score (Z = -2.828, -2.022, -2.015, -2.015, all P < 0.05). The hepatic vein diameter, superior mesenteric vein inner diameter and wall thickness of the ascending colon in the study group were significantly higher than those in the control group (t = 9.543, P < 0.001; t = 2.277, P = 0.025; Z = -2.062, P = 0.039). Antral contraction amplitude, antral contraction frequency, motility index, jejunal peristalsis frequency, and ascending colon peristalsis frequency were significantly lower in the study group compared to the control group (Z = -2.571, -4.196, -3.681, -5.451, -4.061, all P < 0.001). The wall thickness of the antrum, jejunum, and ascending colon were positively correlated with the diameter of the hepatic vein (r = 0.394, P = 0.011; r = 0.352, P = 0.024; r = 0.450, P = 0.003). Motility index and ascending colon peristalsis frequency were positively correlated with the peak velocity of superior mesenteric vein (r = 0.456, P = 0.029; r = 0.507, P = 0.007). The wall thickness of the jejunum was positively correlated with the peak velocity of superior mesenteric artery (r = 0.330, P = 0.035). Peak velocity of superior mesenteric artery, antral contraction frequency, and jejunal peristalsis frequency were negatively correlated with the reflux score (r = -0.409, P = 0.038; r = -0.423, P = 0.032; r = -0.409, P = 0.038). The wall thickness of the ascending colon was positively correlated with the reflux score (r = 0.414, P = 0.035). Conclusion This study found that patients with acute heart failure exhibited thickening of the gastrointestinal wall and generally reduced gastrointestinal motility, with predominantly lower abdominal symptoms. These findings indicate that ultrasound can effectively monitor the gastrointestinal structure and function of patients with acute heart failure, which is expected to provide help for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ruyi Hao
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ye Zheng
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qing Zhao
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jie Chen
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ruiqi Fan
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Peng Chen
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Na Yin
- Department of Emergency, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huai Qin
- Department of Diagnostic Ultrasound, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Nie HY, Ge J, Huang GX, Liu KG, Yue Y, Li H, Lin HG, Zhang T, Yan HF, Xu BX, Sun HW, Yang JW, Si SY, Zhou JL, Cui Y. New insights into the intestinal barrier through "gut-organ" axes and a glimpse of the microgravity's effects on intestinal barrier. Front Physiol 2024; 15:1465649. [PMID: 39450142 PMCID: PMC11499591 DOI: 10.3389/fphys.2024.1465649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/02/2024] [Indexed: 10/26/2024] Open
Abstract
Gut serves as the largest interface between humans and the environment, playing a crucial role in nutrient absorption and protection against harmful substances. The intestinal barrier acts as the initial defense mechanism against non-specific infections, with its integrity directly impacting the homeostasis and health of the human body. The primary factor attributed to the impairment of the intestinal barrier in previous studies has always centered on the gastrointestinal tract itself. In recent years, the concept of the "gut-organ" axis has gained significant popularity, revealing a profound interconnection between the gut and other organs. It speculates that disruption of these axes plays a crucial role in the pathogenesis and progression of intestinal barrier damage. The evaluation of intestinal barrier function and detection of enterogenic endotoxins can serve as "detecting agents" for identifying early functional alterations in the heart, kidney, and liver, thereby facilitating timely intervention in the disorders. Simultaneously, consolidating intestinal barrier integrity may also present a potential therapeutic approach to attenuate damage in other organs. Studies have demonstrated that diverse signaling pathways and their corresponding key molecules are extensively involved in the pathophysiological regulation of the intestinal barrier. Aberrant activation of these signaling pathways and dysregulated expression of key molecules play a pivotal role in the process of intestinal barrier impairment. Microgravity, being the predominant characteristic of space, can potentially exert a significant influence on diverse intestinal barriers. We will discuss the interaction between the "gut-organ" axes and intestinal barrier damage, further elucidate the signaling pathways underlying intestinal barrier damage, and summarize alterations in various components of the intestinal barrier under microgravity. This review aims to offer a novel perspective for comprehending the etiology and molecular mechanisms of intestinal barrier injury as well as the prevention and management of intestinal barrier injury under microgravity environment.
Collapse
Affiliation(s)
- Hong-Yun Nie
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jun Ge
- Clinical laboratory, The Ninth Medical Center of the PLA General Hospital, Beijing, China
| | - Guo-Xing Huang
- 306th Clinical College of PLA, The Fifth Clinical College, Anhui Medical University, Beijing, China
| | - Kai-Ge Liu
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Yuan Yue
- Department of Disease Control and Prevention, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hao Li
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hai-Guan Lin
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Tao Zhang
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hong-Feng Yan
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Bing-Xin Xu
- Special Medical Laboratory Center, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hong-Wei Sun
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jian-Wu Yang
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Shao-Yan Si
- Special Medical Laboratory Center, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jin-Lian Zhou
- Department of Pathology, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Yan Cui
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
29
|
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
|
30
|
Cuervo L, McAlpine PL, Olano C, Fernández J, Lombó F. Low-Molecular-Weight Compounds Produced by the Intestinal Microbiota and Cardiovascular Disease. Int J Mol Sci 2024; 25:10397. [PMID: 39408727 PMCID: PMC11477366 DOI: 10.3390/ijms251910397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is the main cause of mortality in industrialized countries, with over 500 million people affected worldwide. In this work, the roles of low-molecular-weight metabolites originating from the gut microbiome, such as short-chain fatty acids, hydrogen sulfide, trimethylamine, phenylacetic acid, secondary bile acids, indoles, different gases, neurotransmitters, vitamins, and complex lipids, are discussed in relation to their CVD-promoting or preventing activities. Molecules of mixed microbial and human hepatic origin, such as trimethylamine N-oxide and phenylacetylglutamine, are also presented. Finally, dietary agents with cardioprotective effects, such as probiotics, prebiotics, mono- and poly-unsaturated fatty acids, carotenoids, and polyphenols, are also discussed. A special emphasis is given to their gut microbiota-modulating properties.
Collapse
Affiliation(s)
- Lorena Cuervo
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Patrick L. McAlpine
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos Olano
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Javier Fernández
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Felipe Lombó
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
31
|
Sarkar S, Prasanna VS, Das P, Suzuki H, Fujihara K, Kodama S, Sone H, Sreedhar R, Velayutham R, Watanabe K, Arumugam S. The onset and the development of cardiometabolic aging: an insight into the underlying mechanisms. Front Pharmacol 2024; 15:1447890. [PMID: 39391689 PMCID: PMC11464448 DOI: 10.3389/fphar.2024.1447890] [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: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024] Open
Abstract
Metabolic compromise is crucial in aggravating age-associated chronic inflammation, oxidative stress, mitochondrial damage, increased LDL and triglycerides, and elevated blood pressure. Excessive adiposity, hyperglycemia, and insulin resistance due to aging are associated with elevated levels of damaging free radicals, inducing a proinflammatory state and hampering immune cell activity, leading to a malfunctioning cardiometabolic condition. The age-associated oxidative load and redox imbalance are contributing factors for cardiometabolic morbidities via vascular remodelling and endothelial damage. Recent evidence has claimed the importance of gut microbiota in maintaining regular metabolic activity, which declines with chronological aging and cardiometabolic comorbidities. Genetic mutations, polymorphic changes, and environmental factors strongly correlate with increased vulnerability to aberrant cardiometabolic changes by affecting key physiological pathways. Numerous studies have reported a robust link between biological aging and cardiometabolic dysfunction. This review outlines the scientific evidence exploring potential mechanisms behind the onset and development of cardiovascular and metabolic issues, particularly exacerbated with aging.
Collapse
Affiliation(s)
- Sulogna Sarkar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Vani S. Prasanna
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Pamelika Das
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Hiroshi Suzuki
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoru Kodama
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Remya Sreedhar
- School of Pharmacy, Sister Nivedita University, Kolkata, West Bengal, India
| | - Ravichandiran Velayutham
- Director, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| | - Kenichi Watanabe
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, West Bengal, India
| |
Collapse
|
32
|
Fang Z, Zang Q, Chen J, Li Z, Yang D, Wu C, Yang H, Guo N. Whole-body mass spectrometry imaging reveals the systemic metabolic disorder and catecholamines biosynthesis alteration on heart-gut axis in heart failure rat. J Adv Res 2024:S2090-1232(24)00385-0. [PMID: 39270978 DOI: 10.1016/j.jare.2024.09.001] [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: 06/27/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
INTRODUCTION Heart failure (HF) is a systemic metabolic disorder disease, across multiorgan investigations advancing knowledge of progression and treatment of HF. Whole-body MSI provides spatiotemporal information of metabolites in multiorgan and is expected to be a potent tool to dig out the complex mechanism of HF. OBJECTIVES This study aimed at exploring the systemic metabolic disorder in multiorgan and catecholamines biosynthesis alteration on heart-gut axis after HF. METHODS Whole-body MSI was used to characterize metabolic disorder of the whole rat body after HF. An integrated method by MSI, LC-MS/MS and ELISA was utilized to analyze key metabolites and enzymes on heart, small intestine, cecum and colon tissues of rat. Gut microbiota dysbiosis was investigated by 16S rDNA sequencing and metagenomic sequencing. Validation experiments and in vitro experiments were performed to verify the effect of catecholamines biosynthesis alteration on heart-gut axis after HF. RESULTS Whole-body MSI exhibited varieties of metabolites alteration in multiple organs. Remarkably, catecholamine biosynthesis was significantly altered in the serum, heart and intestines of rats. Furthermore, catecholamines and tyrosine hydroxylase were obviously upregulated in heart and colon tissue. Turicibacter_sanguinis was relevant to catecholamines of heart and colon. Validation experiments demonstrated excessive norepinephrine induced cardio-intestinal injury, including significantly elevating the levels of BNP, pro-BNP, LPS, DAO, and increased the abundance of Turicibacter_sanguinis. These alterations could be reversed by metoprolol treatment blocking the effect of norepinephrine. Additionally, in vitro studies demonstrated that norepinephrine promoted the growth of Turicibacter_sanguinis and Turicibacter_sanguinis could import and metabolize norepinephrine. Collectively, excessive norepinephrine exerted bidirectional effects on cardio-intestinal function to participate in the progression of HF. CONCLUSION Our study provides a new approach to elucidate multiorgan metabolic disorder and proposes new insights into heart-gut axis in HF development.
Collapse
Affiliation(s)
- Zhengyu Fang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingce Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiemei Chen
- Department of Pharmacy, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zeyu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
33
|
Tuerhongjiang G, Guo M, Qiao X, Liu J, Xi W, Wei Y, Liu P, Lou B, Wang C, Sun L, Yuan X, Liu H, Xiong Y, Ma Y, Li H, Zhou B, Li L, Yuan Z, Wu Y, She J. Gut Microbiota Regulate Saturated Free Fatty Acid Metabolism in Heart Failure. SMALL SCIENCE 2024; 4:2300337. [PMID: 40212081 PMCID: PMC11935106 DOI: 10.1002/smsc.202300337] [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: 12/10/2023] [Revised: 05/22/2024] [Indexed: 04/13/2025] Open
Abstract
AIMS Heart failure (HF) is associated with profound changes in cardiac metabolism. At present, there is still a lack of relevant research to explore the key microbiome and their metabolites affecting the progression of HF. Herein, the interaction of gut microbiota and circulating free fatty acid (FFA) in HF patients and mice is investigated. METHODS AND RESULTS In HF patients, by applying metagenomics analysis and targeted FFA metabolomics, enriched abundance of Clostridium sporogenes (C.sp) in early and late stage of HF patients, which negatively correlated to saturated free fatty acid (SFA) levels, is identified. KEGG analysis further indicates microbiota gene enrichment in FFA degradation in early HF, and decreased gene expression in FFA synthesis in late HF. In HF mice (C57BL/6J) induced by isoproterenol (ISO), impaired intestinal permeability is observed, and decreased fecal C.sp and increased SFA are further validated. At last, by supplementing C.sp to ISO-induced HF mice, the cardiac function, fibrosis, and myocardial size are partially rescued, together with decreased circulating SFA levels. CONCLUSIONS Clostridium abundance is increased in HF, compensating cardiac function deterioration via downregulation of circulating SFA levels. The results demonstrate that the gut microbiota-SFA axis plays an important role in HF protection, which may provide a strategic advantage for the probiotic therapy development in HF.
Collapse
Affiliation(s)
- Gulinigaer Tuerhongjiang
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Manyun Guo
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Xiangrui Qiao
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Junhui Liu
- Diagnostic DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Wen Xi
- Diagnostic DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Yuanyuan Wei
- Department of CardiologySecond Affiliated HospitalZhejiang UniversitySchool of MedicineHangzhou310058China
| | - Peining Liu
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Bowen Lou
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Chen Wang
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Lizhe Sun
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Xiao Yuan
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Hui Liu
- BiobankFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Ying Xiong
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Yunlong Ma
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Hongbing Li
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Bo Zhou
- Respiratory DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Lijuan Li
- Cardiovascular DepartmentWuzhong People's HospitalNingxia215128China
| | - Zuyi Yuan
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Yue Wu
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| | - Jianqing She
- Cardiovascular DepartmentFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
- Key Laboratory of Environment and Genes Related to DiseasesMinistry of EducationXi'anShaanxi710061China
| |
Collapse
|
34
|
Yuzefpolskaya M, Bohn B, Ladanyi A, Pinsino A, Braghieri L, Carey MR, Clerkin K, Sayer GT, Latif F, Koji T, Uriel N, Nandakumar R, Uhlemann AC, Colombo PC, Demmer RT. Alterations in the sarcopenia index are associated with inflammation, gut, and oral microbiota among heart failure, left ventricular assist device, and heart transplant patients. J Heart Lung Transplant 2024; 43:1395-1408. [PMID: 38744352 DOI: 10.1016/j.healun.2024.04.069] [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: 11/09/2023] [Revised: 03/28/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Sarcopenia, characterized by loss of muscle mass and function, is prevalent in heart failure (HF) and predicts poor outcomes. We investigated alterations in sarcopenia index (SI), a surrogate for skeletal muscle mass, in HF, left ventricular assist device (LVAD), and heart transplant (HT), and assessed its relationship with inflammation and digestive tract (gut and oral) microbiota. METHODS We enrolled 460 HF, LVAD, and HT patients. Repeated measures pre/post-procedures were obtained prospectively in a subset of LVAD and HT patients. SI (serum creatinine/cystatin C) and inflammatory biomarkers (C-reactive protein, interleukin-6, tumor necrosis factor-alpha) were measured in 271 and 622 blood samples, respectively. Gut and saliva microbiota were assessed via 16S ribosomal ribonucleic acid sequencing among 335 stool and 341 saliva samples. Multivariable regression assessed the relationship between SI and (1) New York Heart Association class; (2) pre- versus post-LVAD or HT; and (3) biomarkers of inflammation and microbial diversity. RESULTS Median (interquartile range) natural logarithm (ln)-SI was -0.13 (-0.32, 0.05). Ln-SI decreased across worsening HF class, further declined at 1 month after LVAD and HT, and rebounded over time. Ln-SI was correlated with inflammation (r = -0.28, p < 0.01), gut (r = 0.28, p < 0.01), and oral microbial diversity (r = 0.24, p < 0.01). These associations remained significant after multivariable adjustment in the combined cohort but not for all individual cohorts. The presence of the gut taxa Roseburia inulinivorans was associated with increased SI. CONCLUSIONS SI levels decreased in symptomatic HF and remained decreased long-term after LVAD and HT. In the combined cohort, SI levels covaried with inflammation in a similar fashion and were significantly related to overall microbial (gut and oral) diversity, including specific taxa compositional changes.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York.
| | - Bruno Bohn
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Annamaria Ladanyi
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Alberto Pinsino
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Lorenzo Braghieri
- Division of Cardiovascular Medicine, Department of Cardiology, Cleveland Clinic, Cleveland, Ohio
| | - Matthew R Carey
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Kevin Clerkin
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Gabriel T Sayer
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Farhana Latif
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Takeda Koji
- Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Nir Uriel
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York
| | - Anne-Catrin Uhlemann
- Division of Infectious Diseases and Microbiome and Pathogen Genomics Core, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Paolo C Colombo
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota; Division of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
35
|
Ribitsch W, Lehner TA, Sauseng N, Rosenkranz AR, Schneditz D. Susceptibility of hepato-splanchnic perfusion to intra-abdominal pressure in peritoneal dialysis patients. Perit Dial Int 2024:8968608241275922. [PMID: 39196595 DOI: 10.1177/08968608241275922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND The impact of peritoneal filling on hepato-splanchnic perfusion during peritoneal dialysis has not been fully elucidated yet. METHODS Measurements were done in 20 prevalent peritoneal dialysis patients during a peritoneal equilibration test (PET) with 2L of standard dialysate. Data were obtained in the drained state at baseline (T0), after instillation (T1), and after 2 h of dwell time (T2). Intra-abdominal pressure (IAP) was measured by Durand's approach. The hepatic clearance index (KI) of indocyanine-green (ICG) was determined as an indirect measure of hepato-splanchnic blood flow. Cardiac index (CI), heart rate (HR), and total peripheral resistance index (TPRI) were derived from continuous arterial pulse analysis. Fluid volume overload (VO) was evaluated by multifrequency bioimpedance analysis. Ejection fraction (EF) was obtained from echocardiographic examination. RESULTS IAP was 5.8 ± 3.5 mmHg at baseline (T0), rose to 9.4 ± 2.8 mmHg after instillation of dialysate (T1), and further to 9.7 ± 2.8 mmHg after 2 h of dwell time (p < 0.001). KI slightly declined from 0.60 ± 0.22 L/min/m2 at T0 to 0.53 ± 0.15 L/min/m2 at T1 (p = 0.075), and returned to 0.59 ± 0.22 L/min/m2 at T2 (p = 0.052). CI, HR, and TPRI did not change significantly. In five patients with an EF < 40% KI was significantly lower at T1 (0.42 ± 0.12 L/min/m2; p = 0.039), and further decreased at T2 (0.40 ± 0.04 L/min/m2; p = 0.016) compared to patients with normal EF (T1: 0.58 ± 0.15 L/min/m2 and T2: 0.67 ± 0.22 L/min/m2). CONCLUSIONS Overall, hepatic clearance of ICG as a marker of hepato-splanchnic blood flow is not affected by the filling of the peritoneal cavity.
Collapse
Affiliation(s)
- Werner Ribitsch
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Thomas A Lehner
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Notburga Sauseng
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Alexander R Rosenkranz
- Clinical Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Daniel Schneditz
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| |
Collapse
|
36
|
Fenski M, Abazi E, Gröschel J, Hadler T, Kappelmayer D, Kolligs F, Prieto C, Botnar R, Kunze KP, Schulz-Menger J. Cardiovascular magnetic resonance reveals myocardial involvement in patients with active stage of inflammatory bowel disease. Clin Res Cardiol 2024:10.1007/s00392-024-02503-5. [PMID: 39102000 DOI: 10.1007/s00392-024-02503-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Active inflammatory bowel disease (A-IBD) but not remission (R-IBD) has been associated with an increased risk of cardiovascular death and hospitalization for heart failure. OBJECTIVES Using cardiovascular magnetic resonance (CMR), this study aims to assess adverse myocardial remodeling in patients with IBD in correlation with disease activity. METHODS Forty-four IBD patients without cardiovascular disease (24 female, median-age: 39.5 years, 26 A-IBD, 18 R-IBD) and 44 matched healthy volunteers (HV) were prospectively enrolled. The disease stage was determined by endoscopic and patient-reported criteria. Participants underwent CMR for cardiac phenotyping: cine imaging and strain analysis were performed to assess ventricular function. T1 mapping, extracellular volume and late-gadolinium enhanced images were obtained to assess focal and diffuse myocardial fibrosis. Simultaneous T1 and T2 elevation (T1 > 1049.3 ms, T2 > 54 ms) was considered to indicate a myocardial segment was inflamed. RESULTS 16/44 (16.4%) IBD patients described dyspnea on exertion and 10/44 (22.7%) reported chest pain. A-IBD patients showed impaired ventricular function, indicated by reduced global circumferential and radial strain despite preserved left-ventricular ejection fraction. 16% of all IBD patients had focal fibrosis in a non-ischemic pattern. A-IDB patients had increased markers of diffuse left ventricular fibrosis (T1-values: A-IBD: 1022.0 ± 34.83 ms, R-IBD: 1010.10 ± 32.88 ms, HV: 990.61 ± 29.35 ms, p < .01). Significantly more participants with A-IDB (8/26, 30.8%) had at least one inflamed myocardial segment than patients in remission (0/18) and HV (1/44, 2.3%, p < .01). Markers of diffuse fibrosis correlated with disease activity. CONCLUSION This study, using CMR, provides evidence of myocardial involvement and patterns of adverse left ventricular remodeling in patients with IBD. CLINICAL TRIAL REGISTRATION ISRCTN30941346.
Collapse
Affiliation(s)
- Maximilian Fenski
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research CenterMax-Delbrück Center for Molecular MedicineDepartment of Cardiology and Nephrology, Charité Medical Faculty, HELIOS Klinikum Berlin Buch, Charité - Universitätsmedizin Berlin Lindenberger Weg 80, 13125, Berlin, Germany
| | - Endri Abazi
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research CenterMax-Delbrück Center for Molecular MedicineDepartment of Cardiology and Nephrology, Charité Medical Faculty, HELIOS Klinikum Berlin Buch, Charité - Universitätsmedizin Berlin Lindenberger Weg 80, 13125, Berlin, Germany
| | - Jan Gröschel
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research CenterMax-Delbrück Center for Molecular MedicineDepartment of Cardiology and Nephrology, Charité Medical Faculty, HELIOS Klinikum Berlin Buch, Charité - Universitätsmedizin Berlin Lindenberger Weg 80, 13125, Berlin, Germany
| | - Thomas Hadler
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research CenterMax-Delbrück Center for Molecular MedicineDepartment of Cardiology and Nephrology, Charité Medical Faculty, HELIOS Klinikum Berlin Buch, Charité - Universitätsmedizin Berlin Lindenberger Weg 80, 13125, Berlin, Germany
| | - Diane Kappelmayer
- Department of Internal Medicine and Gastroenterology, HELIOS Klinikum Berlin Buch, Berlin, Germany
| | - Frank Kolligs
- Department of Internal Medicine and Gastroenterology, HELIOS Klinikum Berlin Buch, Berlin, Germany
| | - Claudia Prieto
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rene Botnar
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karl-Philipp Kunze
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- MR Research Collaborations, Siemens Healthcare Limited, Camberley, UK
| | - Jeanette Schulz-Menger
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research CenterMax-Delbrück Center for Molecular MedicineDepartment of Cardiology and Nephrology, Charité Medical Faculty, HELIOS Klinikum Berlin Buch, Charité - Universitätsmedizin Berlin Lindenberger Weg 80, 13125, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
37
|
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
|
38
|
Mejda Z, Alaa N, Ahlem B, Mohamed Hichem L, Arwa G, Imen J, Ben Chaabene N, Abdelfattah Z, Leila S. Acute ischemic colitis complicating an exacerbation of chronic obstructive pulmonary disease: A case report of gut-lung crosstalk. SAGE Open Med Case Rep 2024; 12:2050313X241256862. [PMID: 38812834 PMCID: PMC11135070 DOI: 10.1177/2050313x241256862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
Acute ischemic colitis is a pathology as frequent as it is serious and requires urgent management. It's often occurring in a context of particular thromboembolic or hypovolemic risk, but certain clinical situations are not commonly known to provide mesenteric ischemia. Herein, we report the case of a 47-year-old man who presented with a severe acute colitis occurring in the course of acute exacerbation of a chronic obstructive pulmonary diseases with maintained stability of hemodynamic state. The diagnosis of acute ischemic colitis complicating an exacerbation of chronic obstructive pulmonary diseases was made. A clinical and biological improvement quickly marked the patient's condition after the management of the respiratory problem.
Collapse
Affiliation(s)
- Zakhama Mejda
- Gastroenterology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Nciri Alaa
- Gastroenterology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Bellalah Ahlem
- Cytopathology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | | | - Guediche Arwa
- Gastroenterology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Jemni Imen
- Gastroenterology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Nabil Ben Chaabene
- Gastroenterology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Zakhama Abdelfattah
- Cytopathology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Safer Leila
- Cytopathology Department, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| |
Collapse
|
39
|
Bao M, Li H, Li J. Circulating trimethylamine N-oxide is correlated with high coronary artery atherosclerotic burden in individuals with newly diagnosed coronary heart disease. BMC Cardiovasc Disord 2024; 24:265. [PMID: 38773380 PMCID: PMC11106919 DOI: 10.1186/s12872-024-03937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is a metabolite derived from the gut microbiota and has been reported to be correlated with cardiovascular diseases. Although TMAO is associated with the severity of coronary artery disease in subjects with coronary heart disease (CHD) history. However, the correlation between TMAO and the atherosclerotic burden in newly diagnosed cases of CHD is unknown. METHODS In this hospital-based study, we enrolled 429 individuals newly diagnosed with CHD undergoing coronary angiography. Plasma TMAO was assessed before coronary angiography. SYNTAX score was computed during coronary angiography to estimate the coronary artery atherosclerotic burden. Both linear and logistic regression analyses were conducted to explore the correlation between plasma TMAO levels and SYNTAX score in newly diagnosed CHD population. RESULTS The TMAO in patients with SYNTAX ≥ 33 and subjects with SYNTAX < 23 were 6.10 (interquartile range [IQR]: 3.53 to 9.15) µmol/L and 4.90 [IQR: 3.25 to 7.68] µmol/L, respectively. Linear regression adjusting for traditional risk factors showed TMAO level was positively correlated with SYNTAX score (β = 0.179; p = 0.006) in CHD population. When TMAO was added to models with traditional risk factors, the predictive value improved significantly, with the receiver operating characteristic curve (AUC) increased from 0.7312 to 0.7502 (p = 0.003). Stratified analysis showed that the correlations did not hold true for subjects who were non-smoker or with histories of diabetes. None of the stratifying factors significantly altered the correlation (all p for interaction < 0.05). CONCLUSIONS We found a positive linear correlation between plasma TMAO and SYNTAX score among newly diagnosed CHD individuals in Chinese population.
Collapse
Affiliation(s)
- Minghui Bao
- Department of Cardiology, Peking University First Hospital, Peking University, Xi Shiku Street No. 8, Xicheng District, Beijing, 100034, China.
| | - Haotong Li
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Peking University, Xi Shiku Street No. 8, Xicheng District, Beijing, 100034, China
| |
Collapse
|
40
|
Huang C, Li X, Li H, Chen R, Li Z, Li D, Xu X, Zhang G, Qin L, Li B, Chu XM. Role of gut microbiota in doxorubicin-induced cardiotoxicity: from pathogenesis to related interventions. J Transl Med 2024; 22:433. [PMID: 38720361 PMCID: PMC11077873 DOI: 10.1186/s12967-024-05232-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
Doxorubicin (DOX) is a broad-spectrum and highly efficient anticancer agent, but its clinical implication is limited by lethal cardiotoxicity. Growing evidences have shown that alterations in intestinal microbial composition and function, namely dysbiosis, are closely linked to the progression of DOX-induced cardiotoxicity (DIC) through regulating the gut-microbiota-heart (GMH) axis. The role of gut microbiota and its metabolites in DIC, however, is largely unelucidated. Our review will focus on the potential mechanism between gut microbiota dysbiosis and DIC, so as to provide novel insights into the pathophysiology of DIC. Furthermore, we summarize the underlying interventions of microbial-targeted therapeutics in DIC, encompassing dietary interventions, fecal microbiota transplantation (FMT), probiotics, antibiotics, and natural phytochemicals. Given the emergence of microbial investigation in DIC, finally we aim to point out a novel direction for future research and clinical intervention of DIC, which may be helpful for the DIC patients.
Collapse
Affiliation(s)
- Chao Huang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Xiaoxia Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, No. 308 Ningxia Road, Qingdao, Shandong, 266000, China
| | - Hanqing Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China
| | - Ruolan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Zhaoqing Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Xiaojian Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Guoliang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Luning Qin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, No. 308 Ningxia Road, Qingdao, Shandong, 266000, China.
- Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao, 266033, China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, Shandong, 266100, China.
- The Affiliated Cardiovascular Hospital of Qingdao University, No. 5 Zhiquan Road, Qingdao, 266071, China.
| |
Collapse
|
41
|
Frederiks P, Peetermans M, Wilmer A. Nutritional support in the cardiac intensive care unit. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2024; 13:373-379. [PMID: 38333990 DOI: 10.1093/ehjacc/zuae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/10/2024]
Abstract
Optimal care of critically ill patients in the cardiac intensive care unit includes adequate nutritional support. This review highlights the high prevalence of malnutrition in acute heart failure, acute coronary syndrome, cardiogenic shock, and post-cardiac arrest and its adverse impact on prognosis. There is a lack of robust evidence regarding appropriate nutritional support in this patient population. Initiation of nutritional support with a comprehensive assessment of the patient's nutritional status is critical. High-risk cardiac patients who are not critically ill can receive oral nutrition adapted to individual risk factors or deficiencies, although overfeeding should be avoided in the acute phase. For critically ill patients at risk of or with malnutrition on admission, general principles include initiation of nutritional support within 48 h of admission, preference for enteral over parenteral nutrition, preference for hypocaloric nutrition in the first week of intensive care unit admission, and adequate micronutrient supplementation. Enteral nutrition in haemodynamically unstable patients carries a risk, albeit low, of intestinal ischaemia. In the case of malnutrition, the risk of refeeding syndrome should always be considered.
Collapse
Affiliation(s)
- Pascal Frederiks
- Department of Cardiovascular Medicine, University Hospitals Leuven, Leuven, Belgium
- UZ Gasthuisberg Dept. General Internal Medicine, Herestraat 493000, Leuven, Belgium
| | - Marijke Peetermans
- UZ Gasthuisberg Dept. General Internal Medicine, Herestraat 493000, Leuven, Belgium
- Medical Intensive Care Unit, University Hospitals Leuven, Herestraat 49, B 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Alexander Wilmer
- UZ Gasthuisberg Dept. General Internal Medicine, Herestraat 493000, Leuven, Belgium
- Medical Intensive Care Unit, University Hospitals Leuven, Herestraat 49, B 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
42
|
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
|
43
|
Singh H, Kaushal J, Garcia A, Kak V. Clostridium perfringens Empyema: Anaerobic Invasion in an Uncommon Location. Cureus 2024; 16:e60082. [PMID: 38860109 PMCID: PMC11164247 DOI: 10.7759/cureus.60082] [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] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Clostridium perfringens bacteremia arises due to skin inoculation from the external environment or translocation from the gastrointestinal tract. In the event of bacteremia, it tends to colonize in anaerobic environments due to its obligatory anaerobic nature. Its inoculation in the lung, albeit rare, can occur if an anaerobic nidus is created. In the presented case, the patient developed C. perfringens bacteremia andempyema in the area of lung necrosis caused by acute pulmonary embolism. He did not have any history of chest trauma, and the source of bacteremia was deemed to be via gut translocation. The patient was noted to have multiple gastric ulcers on endoscopy and jejunal wall thickening, which likely led to the bacterial translocation into the bloodstream. He underwent video-assisted thoracoscopic surgery-assisted decortication and intravenous antibiotics, eventually leading to clinical improvement. To identify the source of Clostridium in the absence of penetrating trauma, a thorough gastrointestinal evaluation, including a colonoscopy, is warranted to identify the pathology leading to the gastrointestinal translocation.
Collapse
Affiliation(s)
| | | | | | - Vivek Kak
- Infectious Disease, Henry Ford Health System, Jackson, USA
| |
Collapse
|
44
|
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
|
45
|
Zhang Z, Lv T, Wang X, Wu M, Zhang R, Yang X, Fu Y, Liu Z. Role of the microbiota-gut-heart axis between bile acids and cardiovascular disease. Biomed Pharmacother 2024; 174:116567. [PMID: 38583340 DOI: 10.1016/j.biopha.2024.116567] [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: 12/13/2023] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024] Open
Abstract
Bile acid (BA) receptors (e.g., farnesoid X-activated receptor, muscarinic receptor) are expressed in cardiomyocytes, endothelial cells, and vascular smooth muscle cells, indicating the relevance of BAs to cardiovascular disease (CVD). Hydrophobic BAs are cardiotoxic, while hydrophilic BAs are cardioprotective. For example, fetal cardiac insufficiency in maternal intrahepatic cholestasis during pregnancy, and the degree of fetal cardiac abnormality, is closely related to the level of hydrophobic BAs in maternal blood and infant blood. However, ursodeoxycholic acid (the most hydrophilic BA) can reverse/prevent these detrimental effects of increased levels of hydrophobic BAs on the heart. The gut microbiota (GM) and GM metabolites (especially secondary BAs) have crucial roles in hypertension, atherosclerosis, unstable angina, and heart failure. Herein, we describe the relationship between CVD and the GM at the BA level. We combine the concept of the "microbiota-gut-heart axis" (MGHA) and postulate the role and mechanism of BAs in CVD development. In addition, the strategies for treating CVD with BAs under the MGHA are proposed.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, PR China; Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Tingting Lv
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China; Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang, PR China
| | - Xiang Wang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Menglu Wu
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Ruolin Zhang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Xiaopeng Yang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China
| | - Yongping Fu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, PR China.
| | - Zheng Liu
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, PR China.
| |
Collapse
|
46
|
Villela-Torres MDLL, Prado-Uribe MDC, Díaz MÁ, Pablo HQ, Soria-Castro E, Escofet NE, Maldonado CEF, Paniagua R. Effect of High Sodium Intake on Gut Tight Junctions' Structure and Permeability to Bacterial Toxins in a Rat Model of Chronic Kidney Disease. Arch Med Res 2024; 55:102969. [PMID: 38484487 DOI: 10.1016/j.arcmed.2024.102969] [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: 06/20/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 04/22/2024]
Abstract
INTRODUCTION Uremic toxicity changes the gut structure and permeability, allowing bacterial toxins to translocate from the lumen to the blood during chronic kidney failure (CKD). Clinical fluid overload and tissue edema without uremia have similar effects but have not been adequately demonstrated and analyzed in CKD. AIMS To investigate the effect of sodium intake on the plasma concentration of gut-derived uremic toxins, indoxyl sulfate (IS), and p-cresyl sulfate (pCS) and the expression of genes and proteins of epithelial gut tight junctions in a rat model of CKD. METHODS Sham-operated (control group, CG) and five-sixths nephrectomized (5/6Nx) Sprague-Dawley rats were randomly assigned to low (LNa), normal (NNa), or high sodium (HNa) diets., Animals were then sacrificed at 8 and 12 weeks and analyzed for IS and pCS plasma concentrations, as well as for gene and protein expression of thigh junction proteins, and transmission electron microscopy (TEM) in colon fragments. RESULTS The HNa 5/6Nx groups had higher concentrations of IS and pCS than CG, NNa, and LNa at eight and twelve weeks. Furthermore, HNa 5/6Nx groups had reduced expression of the claudin-4 gene and protein than CG, NNa, and LNa. HNa had reduced occludin gene expression compared to CG. Occludin protein expression was more reduced in HNa than in CG, NNa, and LNa. The gut epithelial tight junctions appear dilated in HNa compared to NNa and LNa in TEM. CONCLUSION Dietary sodium intake and fluid overload have a significant role in gut epithelial permeability in the CKD model.
Collapse
Affiliation(s)
- María de la Luz Villela-Torres
- Medical Research Unit in Nephrological Diseases, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - María-Del-Carmen Prado-Uribe
- Medical Research Unit in Nephrological Diseases, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Marcela Ávila Díaz
- Medical Research Unit in Nephrological Diseases, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Héctor Quezada Pablo
- Immunnology and Proteomics Research Lab, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Elizabeth Soria-Castro
- Cardiovascular Biomedicine Department, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Nuria Esturau Escofet
- Physical Chemistry Department, Laboratorio Universitario de Resonancia Magnética Nuclear, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Ramón Paniagua
- Medical Research Unit in Nephrological Diseases, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| |
Collapse
|
47
|
Zych-Krekora K, Sylwestrzak O, Krekora M, Oszukowski P, Grzesiak M. Potential Impact of a Pregnant Woman's Microbiota on the Development of Fetal Heart Defects: A Review of the Literature. Biomedicines 2024; 12:654. [PMID: 38540267 PMCID: PMC10968164 DOI: 10.3390/biomedicines12030654] [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: 01/11/2024] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 11/11/2024] Open
Abstract
Developments in medicine and biology in recent decades have led to a significant increase in our knowledge of the complex interactions between the microbiota and human health. In the context of perinatal medicine and neonatology, particular attention is being paid to the potential impact of the maternal microbiota on fetal development. Among the many aspects of this relationship, the question of the impact of dysbiosis on the development of fetal heart defects is an important one. In this article, we present an analysis of recent research and scientific evidence on the relationship between a pregnant woman's microbiota and the development of fetal heart defects. We also discuss potential intervention strategies, including the role of probiotics and diet in optimising the maternal microbiota.
Collapse
Affiliation(s)
- Katarzyna Zych-Krekora
- Department of Perinatology, Obstetrics and Gynaecology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
| | - Oskar Sylwestrzak
- Department of Obstetrics and Gynaecology, Polish Mother’s Memorial Health Institute, 93-338 Lodz, Poland; (O.S.); (M.K.)
- Department of Prenatal Cardiology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
| | - Michał Krekora
- Department of Obstetrics and Gynaecology, Polish Mother’s Memorial Health Institute, 93-338 Lodz, Poland; (O.S.); (M.K.)
- Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-419 Lodz, Poland
| | - Przemysław Oszukowski
- Obstetrics Ward of the Polish Mother’s Memorial Health Institute, 93-338 Lodz, Poland;
- Department of Obstetrics, Gynaecology and Gynaecology Oncology, Medical University of Lodz, 93-113 Lodz, Poland
| | - Mariusz Grzesiak
- Department of Perinatology, Obstetrics and Gynaecology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
- Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
48
|
Luxford JC, Casey CE, Roberts PA, Irving CA. Iron deficiency and anemia in pediatric dilated cardiomyopathy are associated with clinical, biochemical, and hematological markers of severe disease and adverse outcomes. J Heart Lung Transplant 2024; 43:379-386. [PMID: 38012978 DOI: 10.1016/j.healun.2023.11.014] [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: 06/21/2023] [Revised: 10/21/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND There is limited evidence regarding the prevalence and impact of iron deficiency (ID) in children with dilated cardiomyopathy (DCM). METHODS Retrospective single-center review of all children between 2010 and 2020 with a diagnosis of DCM and complete iron studies. ID was defined as ≥2 of ferritin <20 μg/liter, iron <9 μmol/liter, transferrin >3 g/liter, or transferrin saturation (TSat) <15%. Clinical and laboratory indices and freedom from a composite adverse event (CAE) of mechanical circulatory support (MCS), heart transplant, or death were compared between children with and without ID. RESULTS Of 138 patients with DCM, 47 had available iron studies. Twenty-nine (62%) were iron deficient. Children with ID were more likely to be receiving inotropes (17, 59%, p = 0.005) or invasive/noninvasive ventilation (13, 45%, p = 0.016) than those who were iron replete. They had a higher incidence of anemia (22, 76%, p = 0.004) and higher NT-proBNP (1,590 pmol/liter, IQR 456-3,447, p = 0.001). Children with ID had significantly less freedom from the CAE at 1-year (54% ± 10%), 2-years (45 ± 10), and 5-years (37% ± 11%) than those without (p = 0.011). ID and anemia were the only significant predictors of the CAE on univariate Cox regression. CONCLUSIONS ID is highly prevalent in children with DCM. Iron studies are undermeasured in clinical practice, but ID is associated with severe heart failure (HF) and an increased risk of the CAE. The need for iron replacement therapy should be considered in children who present in HF with DCM.
Collapse
Affiliation(s)
- Jack C Luxford
- Heart Centre for Children, Children's Hospital at Westmead, Sydney, Australia; Childrens Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.
| | - Charlene E Casey
- Heart Centre for Children, Children's Hospital at Westmead, Sydney, Australia
| | - Philip A Roberts
- Heart Centre for Children, Children's Hospital at Westmead, Sydney, Australia
| | - Claire A Irving
- Heart Centre for Children, Children's Hospital at Westmead, Sydney, Australia; Childrens Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| |
Collapse
|
49
|
Hullon D, Taherifard E, Al-Saraireh TH. The effect of the four pharmacological pillars of heart failure on haemoglobin level. Ann Med Surg (Lond) 2024; 86:1575-1583. [PMID: 38463117 PMCID: PMC10923357 DOI: 10.1097/ms9.0000000000001773] [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: 11/28/2023] [Accepted: 01/21/2024] [Indexed: 03/12/2024] Open
Abstract
Anaemia, a condition characterized by low levels of haemoglobin, is frequently observed in patients with heart failure (HF). Guideline-directed medical therapy improves HF outcomes by using medications like beta blockers, angiotensin-converting enzyme inhibitors, and angiotensin receptor blockers, along with mineralocorticoid receptor antagonists and sodium-glucose cotransporter 2 inhibitors. In this study, we aimed to review the pathophysiology of anaemia in patients with HF and present the current evidence regarding the relationship between the main recommended medications for these patients and haemoglobin levels. The authors conducted a comprehensive search in the medical literature for relevant original clinical articles in which the four pharmacological pillars of HF were given to the patients; we, then, assessed whether the association of use of these medications and haemoglobin level or development of anaemia was provided. These common medications have been shown in the literature that may exacerbate or ameliorate anaemia. Besides, it has been shown that even in the case that they result in the development of anaemia, their use is associated with positive effects that outweigh this potential harm. The literature also suggests that among patients receiving medications with negative effects on the level of haemoglobin, there was no difference in the rate of mortality between anaemic and non-anaemic patients when both were on treatment for anaemia; this point highlights the importance of the detection and treatment of anaemia in these patients. Further research is needed to explore these relationships and identify additional strategies to mitigate the risk of anaemia in this population.
Collapse
Affiliation(s)
| | - Erfan Taherifard
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
50
|
Rashid S, Sado AI, Afzal MS, Ahmed A, Almaalouli B, Waheed T, Abid R, Majumder K, Kumar V, Tejwaney U, Kumar S. Role of gut microbiota in cardiovascular diseases - a comprehensive review. Ann Med Surg (Lond) 2024; 86:1483-1489. [PMID: 38463085 PMCID: PMC10923299 DOI: 10.1097/ms9.0000000000001419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/30/2023] [Indexed: 03/12/2024] Open
Abstract
The connection between cardiovascular illnesses and the gut microbiota has drawn more and more attention in recent years. According to research, there are intricate relationships between dietary elements, gut bacteria, and their metabolites that affect cardiovascular health. In this study, the role of gut microbiota in cardiovascular disorders is examined, with an emphasis on the cardiac consequences brought on by changes in gut microbiota. This essay discusses the gut-heart axis in depth and in detail. It talks about clinical research looking at how soy consumption, probiotic supplements, and dietary changes affected gut microbiota and cardiovascular risk variables. Our goal is to clarify the possible pathways that connect gut microbiota to cardiovascular health and the implications for upcoming treatment approaches. The authors examine the composition, roles, and effects of the gut microbiota on cardiovascular health, including their contributions to hypertension, atherosclerosis, lipid metabolism, and heart failure. Endotoxemia, inflammation, immunological dysfunction, and host lipid metabolism are some of the potential processes investigated for how the gut microbiota affects cardiac outcomes. The research emphasizes the need for larger interventional studies and personalized medicine strategies to completely understand the complexity of the gut-heart axis and its implications for the management of cardiovascular disease. The development of novel treatment strategies and cutting-edge diagnostic technologies in cardiovascular medicine may be facilitated by a better understanding of this axis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rabia Abid
- Liaquat college of medicine and dentistry, Karachi, Pakistan
| | | | | | | | - Sarwan Kumar
- Wayne State University
- Department of Medicine, Chittagong Medical College, Chittagong, Bangladesh
| |
Collapse
|