1
|
Niu Y, Xiao L, Feng L. Association between dietary index for gut microbiota and metabolic syndrome risk: a cross-sectional analysis of NHANES 2007-2018. Sci Rep 2025; 15:15153. [PMID: 40307409 PMCID: PMC12044051 DOI: 10.1038/s41598-025-99396-9] [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: 01/21/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025] Open
Abstract
Metabolic syndrome (MetS) poses a significant global health challenge, closely associated with cardiovascular diseases, diabetes, and other conditions. With the global prevalence of MetS steadily rising, the potential role of gut microbiota in its development has garnered increasing attention. Against this backdrop, the present study aims to explore the association between the dietary index for gut microbiota (DI-GM) score and MetS. This cross-sectional study utilized data from the 2007-2018 U.S. National Health and Nutrition Examination Survey (NHANES), including 339,242 adults aged ≥ 18 years. The DI-GM score, constructed based on 14 food or nutrient components, served as the exposure variable. MetS was defined according to the Adult Treatment Panel III (ATP III) criteria, including abdominal obesity (waist circumference ≥ 102 cm in men and ≥ 88 cm in women), elevated triglycerides (≥ 150 mg/dL), reduced HDL cholesterol (< 40 mg/dL in men and < 50 mg/dL in women), elevated blood pressure (≥ 130/85 mmHg), and elevated fasting glucose (≥ 100 mg/dL). Multivariable logistic regression analyses were performed to adjust for demographic characteristics, lifestyle factors, and other potential confounders. Higher DI-GM scores were significantly associated with a reduced risk of MetS. After adjusting for all confounders, individuals in the highest quartile (Q4) of DI-GM scores had a 16% lower risk of MetS compared to those in the lowest quartile (Q1) (OR: 0.84; 95%CI: 0.70-1.01). Mediation analyses revealed that systemic immune-inflammation index (SII) and neutrophil-to-lymphocyte ratio (NLR) mediated 4.63% and 3.83% of the association between DI-GM and MetS, respectively. There is an inverse association between DI-GM scores and the risk of MetS, potentially mediated in part by inflammatory markers. These findings provide new evidence supporting dietary interventions aimed at improving gut microbiota to prevent MetS.
Collapse
Affiliation(s)
- Yueyue Niu
- the Cadre Health Care Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Xiao
- the Cadre Health Care Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ling Feng
- the Cadre Health Care Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Tonon CR, Pereira AG, Ferreira NF, Monte MG, Vieira NM, Fujimori ASS, Ballin PDS, de Paiva SAR, Zornoff LAM, Minicucci MF, Polegato BF. The Gut-Heart Axis and Its Role in Doxorubicin-Induced Cardiotoxicity: A Narrative Review. Microorganisms 2025; 13:855. [PMID: 40284691 PMCID: PMC12029146 DOI: 10.3390/microorganisms13040855] [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: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Doxorubicin is a widely used chemotherapy for the treatment of several types of cancer. However, its application is restricted due to adverse effects, particularly cardiotoxicity, which can progress to heart failure-a chronic and debilitating condition. Several mechanisms have been identified in the pathophysiology of doxorubicin-induced cardiotoxicity, including oxidative stress, mitochondrial dysfunction, inflammation, and disruption of collagen homeostasis. More recently, dysbiosis of the gut microbiota has been implicated in the development and perpetuation of cardiac injury. Studies have reported alterations in the composition and abundance of the microbiota during doxorubicin treatment. Therefore, as of recent, there is a new field of research in order to develop strategies involving the gut microbiota to prevent or attenuate cardiotoxicity since there is no effective therapy at the moment. This narrative review aims to provide an update on the role of gut microbiota and intestinal permeability in the pathophysiology of cardiovascular diseases, and more specifically doxorubicin-induced cardiotoxicity. Additionally, it seeks to establish a foundation for future research targeting gut microbiota to alleviate cardiotoxicity.
Collapse
Affiliation(s)
- Carolina Rodrigues Tonon
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, Brazil (B.F.P.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Yafarova AA, Dementeva EV, Zlobovskaya OA, Sheptulina AF, Lopatukhina EV, Timofeev YS, Glazunova EV, Lyundup AV, Doludin YV, Kiselev AR, Shipulin GA, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota and Metabolic Alterations Associated with Heart Failure and Coronary Artery Disease. Int J Mol Sci 2024; 25:11295. [PMID: 39457077 PMCID: PMC11508380 DOI: 10.3390/ijms252011295] [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: 09/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced ejection fraction (HFrEF). This cross-sectional study included 189 participants divided into three groups: coronary artery disease (n = 93), heart failure with reduced ejection fraction (n = 43), and controls (n = 53). Assessments included physical exams, echocardiography, dietary surveys, blood analysis, and fecal analysis. Gut microbiota composition was analyzed using next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR). Statistical analysis methods for testing hypotheses and correlations, alpha and beta-diversity analyses, co-occurrence networks, and machine learning were conducted using Python libraries or R packages with multiple comparisons corrected using the Benjamini-Hochberg procedure. Significant gut microbiota alterations were observed, with higher Bacillota/Bacteroidota ratios in CAD and HFrEF groups compared to controls (p < 0.001). Significant differences were observed in α-diversity indices (Pielou, Chao1, Faith) between disease groups and controls (p < 0.001). β-diversity analyses also revealed distinct microbial profiles (p = 0.0015). Interestingly, trimethylamine N-oxide (TMAO) levels were lower in CAD and HFrEF groups compared to controls (p < 0.05), while indoxyl sulfate (IS) levels were comparable between the study groups. Co-occurrence network analysis and machine learning identified key microbial features linked to these conditions, highlighting complex interactions within the gut microbiota associated with cardiovascular disease.
Collapse
Affiliation(s)
- Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Dementeva
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Olga A. Zlobovskaya
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Lopatukhina
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Yuriy S. Timofeev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Evgeniya V. Glazunova
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Aleksey V. Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, 117036 Moscow, Russia
| | - Yuriy V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - German A. Shipulin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Valentin V. Makarov
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Sergey M. Yudin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| |
Collapse
|
4
|
Li Z, Xu Q, Huangfu N, Cui H. The effect and mechanism of inulin on atherosclerosis is mediated by the characteristic intestinal flora and metabolites. Coron Artery Dis 2024; 35:498-508. [PMID: 38767579 DOI: 10.1097/mca.0000000000001377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
BACKGROUND Inflammation and hyperlipidemia can cause atherosclerosis. Prebiotic inulin has been proven to effectively reduce inflammation and blood lipid levels. Utilizing a mouse model induced by a high-fat diet, this study aimed to explore whether the characteristic intestinal flora and its metabolites mediate the effects of inulin intervention on atherosclerosis and to clarify the specific mechanism. METHODS Thirty apolipoprotein E-deficient (ApoE-/-) mice were randomly divided into three groups. They were fed with a normal diet, a high-fat diet or an inulin+high-fat diet for 16 weeks. The total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) in the three groups were compared. The gross aorta and aortic sinus of mice were stained with oil red O, and the area of atherosclerotic plaque was observed and compared. The diversity and structure of the mouse fecal flora were detected by sequencing the V3-V4 region of the 16S rRNA gene, and the levels of metabolites in mouse feces were assessed by gas chromatography-mass spectrometry. The plasma lipopolysaccharide (LPS) levels and aortic inflammatory factors were measured by multi-index flow cytometry (CBA). RESULTS ApoE-/- mice fed with the high-fat diet exhibited an increase of approximately 46% in the area of atherosclerotic lesions, and the levels of TC, TG and LDL-C were significantly increased ( P < 0.05) compared with levels in the normal diet group. After inulin was added to the high-fat group, the area of atherosclerotic lesions, the level of serum LPS and aortic inflammation were reduced, and the levels of TC, TG and LDL-C were decreased ( P < 0.05). Based on 16S rRNA gene detection, we found that the composition of the intestinal microbiota, such as Prevotella, and metabolites, such as L-arginine, changed significantly due to hyperlipidemia, and the dietary inulin intervention partially reversed the relevant changes. CONCLUSION Inulin can inhibit the formation of atherosclerotic plaques, which may be related to the changes in lipid metabolism, the composition of the intestinal microbial community and its metabolites, and the inhibition of the expression of related inflammatory factors. Our study identified the relationships among the characteristic intestinal microbiota, metabolites and atherosclerosis, aiming to provide a new direction for future research to delay or treat atherosclerosis by changing the composition and function of the host intestinal microbiota and metabolites.
Collapse
Affiliation(s)
| | - Qingqing Xu
- Department of Nephrology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | | | | |
Collapse
|
5
|
Ferenc K, Marcinkowski M, Olszewski J, Kowalczyk P, Pilžys T, Garbicz D, Dib N, Świderska B, Matyba P, Gajewski Z, Grzesiuk E, Zabielski R. The proteomic profile is altered but not repaired after bariatric surgery in type 2 diabetes pigs. Sci Rep 2024; 14:10235. [PMID: 38702370 PMCID: PMC11068747 DOI: 10.1038/s41598-024-60022-9] [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: 11/14/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
To reveal the sources of obesity and type 2 diabetes (T2D) in humans, animal models, mainly rodents, have been used. Here, we propose a pig model of T2D. Weaned piglets were fed high fat/high sugar diet suppling 150% of metabolizable energy. Measurements of weight gain, blood morphology, glucose plasma levels, cholesterol, and triglycerides, as well as glucose tolerance (oral glucose tolerance test, OGTT) were employed to observe T2D development. The histology and mass spectrometry analyses were made post mortem. Within 6 months, the high fat-high sugar (HFHS) fed pigs showed gradual and significant increase in plasma triglycerides and glucose levels in comparison to the controls. Using OGTT test, we found stable glucose intolerance in 10 out of 14 HFHS pigs. Mass spectrometry analysis indicated significant changes in 330 proteins in the intestine, liver, and pancreas of the HFHS pigs. These pigs showed also an increase in DNA base modifications and elevated level of the ALKBH proteins in the tissues. Six diabetic HFHS pigs underwent Scopinaro bariatric surgery restoring glycaemia one month after surgery. In conclusion, a high energy diet applied to piglets resulted in the development of hyperlipidaemia, hyperglycaemia, and type 2 diabetes being reversed by a bariatric procedure, excluding the proteomic profile utill one month after the surgery.
Collapse
Affiliation(s)
- Karolina Ferenc
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Michał Marcinkowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Jarosław Olszewski
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Paweł Kowalczyk
- Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110, Jabłonna, Poland
| | - Tomaš Pilžys
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Damian Garbicz
- Institute of Oncology, Maria Sklodowska-Curie National Research, W.K. Roentgena 5, 02-781, Warsaw, Poland
| | - Naser Dib
- European Health Centre Otwock (ECZ Otwock), The Fryderyk Chopin Hospital, Borowa 14/18, 05-400, Otwock, Poland
| | - Bianka Świderska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Piotr Matyba
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Zdzisław Gajewski
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Elżbieta Grzesiuk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland.
| | - Romuald Zabielski
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland.
| |
Collapse
|
6
|
Shariff S, Kwan Su Huey A, Parag Soni N, Yahia A, Hammoud D, Nazir A, Uwishema O, Wojtara M. Unlocking the gut-heart axis: exploring the role of gut microbiota in cardiovascular health and disease. Ann Med Surg (Lond) 2024; 86:2752-2758. [PMID: 38694298 PMCID: PMC11060260 DOI: 10.1097/ms9.0000000000001744] [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: 09/23/2023] [Accepted: 12/24/2023] [Indexed: 05/04/2024] Open
Abstract
Introduction Gut microbiota has emerged as a pivotal player in cardiovascular health and disease, extending its influence beyond the gut through intricate metabolic processes and interactions with the immune system. Accumulating evidence supports a significant association between gut microbiota and cardiovascular diseases such as atherosclerosis, hypertension, and heart failure. Dietary patterns have been identified as key factors shaping the composition of the gut microbiota and exerting notable impacts on cardiovascular health. Probiotics and prebiotics have shown promise in mitigating the risks of cardiovascular disease by modulating key cardiovascular parameters. Faecal microbiota transplantation (FMT) has recently emerged as a novel and intriguing therapeutic strategy. Aim This review paper aims to explore and elucidate the multifaceted role of gut microbiota in cardiovascular health. It will also address the prevailing challenges and limitations in gut microbiota studies, emphasizing the importance of future research in overcoming these obstacles to expand our understanding of the gut-heart axis. Materials and methods A comprehensive literature search was conducted using various databases including ClinicalTrials, Google Scholar, PubMed, ScienceDirect, MEDLINE, and Ovid Resources. The search strategy included utilizing keywords such as "Gut microbiota," "Randomized controlled trials (RCTs)," "Gut-heart axis," "Dysbiosis," "Diet," "Probiotics," "Prebiotics," "Faecal Microbiota transplantation," "cardiovascular disease," "Meta-analyses," and other compatible terms thereof. Only articles written in English were considered, and selection criteria included relevance to the research objectives, reasonable sample sizes, and robust methodology. In addition to the identified articles, meta-analyses, animal models and studies, and references from the selected articles were also examined to ensure a comprehensive review of the literature. Results Dietary patterns exert a significant influence on the composition of the gut microbiota, and certain diets, such as the Mediterranean diet, have been associated with a favourable gut microbiota profile and a reduced risk of cardiovascular disease (CVD). Probiotics and prebiotics have emerged as potential interventions to mitigate CVD risks by modulating blood pressure, glycemic control, lipid profiles, and gut dysbiosis. Another innovative therapeutic approach is FMT, which involves transferring faecal material from a healthy donor to restore a balanced gut microbiota. FMT holds promise for improving cardiometabolic parameters in individuals with CVD, although further research is needed to elucidate its precise mechanisms and assess its effectiveness. Conclusion The gut microbiota is emerging as a potential therapeutic target for CVD prevention and management. However, current research has limitations, including the need for larger and more diverse studies, the challenges of establishing causality, and concerns regarding the long-term consequences and safety of gut microbiota modulation. Despite these limitations, understanding the gut-heart axis holds promise for the development of personalized therapies and interventions for cardiovascular health. Further research is needed to expand our knowledge and address the ethical and safety issues associated with gut microbiota modification.
Collapse
Affiliation(s)
- Sanobar Shariff
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - Alicia Kwan Su Huey
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Medical School, University of Glasgow, Glasgow, UK
| | - Nishant Parag Soni
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- GMERS Medical College, Ahmedabad, India
| | - Amer Yahia
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Mohamed 6 of Health Sciences, Casablanca, Morocco
| | - Doha Hammoud
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- National University of Pharmacy, Kharkiv, Ukraine
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Clinton Global Initiative University, New York, NY
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Magda Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- University of Michigan Medical School; Ann Arbor, MI
| |
Collapse
|
7
|
Abrignani V, Salvo A, Pacinella G, Tuttolomondo A. The Mediterranean Diet, Its Microbiome Connections, and Cardiovascular Health: A Narrative Review. Int J Mol Sci 2024; 25:4942. [PMID: 38732161 PMCID: PMC11084172 DOI: 10.3390/ijms25094942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
The Mediterranean diet (MD), rich in minimally processed plant foods and in monounsaturated fats but low in saturated fats, meat, and dairy products, represents one of the most studied diets for cardiovascular health. It has been shown, from both observational and randomized controlled trials, that MD reduces body weight, improves cardiovascular disease surrogates such as waist-to-hip ratios, lipids, and inflammation markers, and even prevents the development of fatal and nonfatal cardiovascular disease, diabetes, obesity, and other diseases. However, it is unclear whether it offers cardiovascular benefits from its individual components or as a whole. Furthermore, limitations in the methodology of studies and meta-analyses have raised some concerns over its potential cardiovascular benefits. MD is also associated with characteristic changes in the intestinal microbiota, mediated through its constituents. These include increased growth of species producing short-chain fatty acids, such as Clostridium leptum and Eubacterium rectale, increased growth of Bifidobacteria, Bacteroides, and Faecalibacterium prausnitzii species, and reduced growth of Firmicutes and Blautia species. Such changes are known to be favorably associated with inflammation, oxidative status, and overall metabolic health. This review will focus on the effects of MD on cardiovascular health through its action on gut microbiota.
Collapse
Affiliation(s)
- Vincenzo Abrignani
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Andrea Salvo
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Gaetano Pacinella
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
8
|
Wang XA, Li JP, Lee MS, Yang SF, Chang YS, Chen L, Li CW, Chao YH. A common trajectory of gut microbiome development during the first month in healthy neonates with limited inter-individual environmental variations. Sci Rep 2024; 14:3264. [PMID: 38332050 PMCID: PMC10853277 DOI: 10.1038/s41598-024-53949-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/07/2024] [Indexed: 02/10/2024] Open
Abstract
The early development of the gut microbiome is governed by multiple factors and has significantly long-term effects on later-in-life health. To minimize inter-individual variations in the environment, we determined developmental trajectories of the gut microbiome in 28 healthy neonates during their stay at a postpartum center. Stool samples were collected at three time points: the first-pass meconium within 24 h of life, and at 7 and 28 days of age. Illumina sequencing of the V3-V4 region of 16S rRNA was used to investigate microbiota profiles. We found that there was a distinct microbiota structure at each time point, with a significant shift during the first week. Proteobacteria was most abundant in the first-pass meconium; Firmicutes and Actinobacteria increased with age and were substituted as the major components. Except for a short-term influence of different delivery modes on the microbiota composition, early microbiome development was not remarkably affected by gravidity, maternal intrapartum antibiotic treatment, premature rupture of membranes, or postnatal phototherapy. Hence, our data showed a similar developmental trajectory of the gut microbiome during the first month in healthy neonates when limited in environmental variations. Environmental factors external to the host were crucial in the early microbiome development.
Collapse
Affiliation(s)
- Xing-An Wang
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, 402, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ju-Pi Li
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Maw-Sheng Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Lee Women's Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Sheng Chang
- Department of Research and Development, AllBio Life Incorporation, Taichung, Taiwan
| | - Ling Chen
- Department of Research and Development, AllBio Life Incorporation, Taichung, Taiwan
| | - Chang-Wei Li
- Department of Research and Development, AllBio Life Incorporation, Taichung, Taiwan
| | - Yu-Hua Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, 402, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
- Department of Clinical Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
9
|
Lenzi A, Biagini D, Ghimenti S, Vivaldi FM, Salvo P, Di Francesco F, Lomonaco T. HiSorb sorptive extraction for determining salivary short chain fatty acids and hydroxy acids in heart failure patients. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1228:123826. [PMID: 37481789 DOI: 10.1016/j.jchromb.2023.123826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
Variations in salivary short-chain fatty acids and hydroxy acids (e.g., lactic acid, and 3-hydroxybutyric acid) levels have been suggested to reflect the dysbiosis of human gut microbiota, which represents an additional factor involved in the onset of heart failure (HF) disease. The physical-chemical properties of these metabolites combined with the complex composition of biological matrices mean that sample pre-treatment procedures are almost unavoidable. This work describes a reliable, simple, and organic solvent free protocol for determining short-chain fatty acids and hydroxy acids in stimulated saliva samples collected from heart failure, obese, and hypertensive patients. The procedure is based on in-situ pentafluorobenzyl bromide (PFB-Br) derivatization and HiSorb sorptive extraction coupled to thermal desorption and gas chromatography-tandem mass spectrometry. The HiSorb extraction device is completely compatible with aqueous matrices, thus saving on time and materials associated with organic solvent-extraction methods. A Central Composite Face-Centred experimental design was used for the optimization of the molar ratio between PFB-Br and target analytes, the derivatization temperature, and the reaction time which were 100, 60 °C, and 180 min, respectively. Detection limits in the range 0.1-100 µM were reached using a small amount of saliva (20 µL). The use of sodium acetate-1-13C as an internal standard improved the intra- and inter-day precision of the method which ranged from 10 to 23%. The optimized protocol was successfully applied for what we believe is the first time to evaluate the salivary levels of short chain fatty acids and hydroxy acids in saliva samples of four groups of patients: i) patients admitted to hospital with acute HF symptoms, ii) patients with chronic HF symptoms, iii) patients without HF symptoms but with obesity, and iv) patients without HF symptoms but with hypertension. The first group of patients showed significantly higher levels of salivary acetic acid and lactic acid at hospital admission as well as the lowest values of hexanoic acid and heptanoic acid. Moreover, the significant high levels of acetic acid, propionic acid, and butyric acid observed in HF respect to the other patients suggest the potential link between oral bacteria and gut dysbiosis.
Collapse
Affiliation(s)
- Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Silvia Ghimenti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Federico M Vivaldi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Pietro Salvo
- Institute of Clinical Physiology, CNR, Via Giuseppe Moruzzi 3, Pisa, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy.
| |
Collapse
|
10
|
Hijová E. Benefits of Biotics for Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24076292. [PMID: 37047262 PMCID: PMC10093891 DOI: 10.3390/ijms24076292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Cardiovascular diseases are the main cause of death in many countries, and the better prevention and prediction of these diseases would be of great importance for individuals and society. Nutrition, the gut microbiota, and metabolism have raised much interest in the field of cardiovascular disease research in the search for the main mechanisms that promote cardiovascular diseases. Understanding the interactions between dietary nutrient intake and the gut microbiota-mediated metabolism may provide clinical insight in order to identify individuals at risk of cardiometabolic disease progression, as well as other potential therapeutic targets to mitigate the risk of cardiometabolic disease progression. The development of cardiometabolic diseases can be modulated by specific beneficial metabolites derived from bacteria. Therefore, it is very important to investigate the impact of these metabolites on human health and the possibilities of modulating their production with dietary supplements called biotics.
Collapse
Affiliation(s)
- Emília Hijová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 040 11 Košice, Slovakia
| |
Collapse
|
11
|
Yuzefpolskaya M, Bohn B, Ladanyi A, Khoruts A, Colombo PC, Demmer RT. Oral and gut microbiome alterations in heart failure: Epidemiology, pathogenesis and response to advanced heart failure therapies. J Heart Lung Transplant 2023; 42:291-300. [PMID: 36586790 DOI: 10.1016/j.healun.2022.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Despite significant advances in therapies, heart failure (HF) remains a progressive disease that, once advanced, is associated with significant death and disability. Cardiac replacement therapies with left ventricular assist device (LVAD) and heart transplantation (HT) are the only treatment options for advanced HF, while lifesaving they can also be lifespan limiting due to the associated complications. Systemic inflammation is mechanistically important in HF pathophysiology and progression. However, directly targeting inflammation in HF has not been beneficial thus far. These failed attempts at therapeutics might be related to our limited understanding of the factors that cause inflammation in HF, and, therefore, to our inability to investigate these triggers in interventional studies. Observational studies have consistently demonstrated associations between alterations in the digestive (gut and oral) microbiome, inflammation and HF risk and progression. Additionally, recent data indicate that these microbial perturbations persist following LVAD and HT, along with residual inflammation and oxidative stress. Furthermore, there is rising recognition of the critical contribution of the microbiome to the metabolism of immunosuppressive drugs after HT. Cumulatively, these findings might posit a mechanistic link between microbiome alterations, systemic inflammation, and adverse outcomes in HF patients before and after cardiac replacement therapies. This review (1) provides an update on available data linking changes in digestive tract microbiota, inflammation, and oxidative stress, to HF pathogenesis and progression; (2) describes evolution of these relationships following LVAD and HT; and (3) outlines present and future intervention strategies that can manipulate the microbiome and possibly modify HF disease trajectory.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, 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, Columbia University Irving Medical Center, New York City, New York
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine University of Minnesota, Minneapolis, Minnesota
| | - Paolo C Colombo
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, 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
|
12
|
Lee JH, Woo KJ, Hong J, Han KI, Kim HS, Kim TJ. Heat-Killed Enterococcus faecalis Inhibit FL83B Hepatic Lipid Accumulation and High Fat Diet-Induced Fatty Liver Damage in Rats by Activating Lipolysis through the Regulation the AMPK Signaling Pathway. Int J Mol Sci 2023; 24:ijms24054486. [PMID: 36901915 PMCID: PMC10002555 DOI: 10.3390/ijms24054486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Continuous consumption of high-calorie meals causes lipid accumulation in the liver and liver damage, leading to non-alcoholic fatty liver disease (NAFLD). A case study of the hepatic lipid accumulation model is needed to identify the mechanisms underlying lipid metabolism in the liver. In this study, the prevention mechanism of lipid accumulation in the liver of Enterococcus faecalis 2001 (EF-2001) was extended using FL83B cells (FL83Bs) and high-fat diet (HFD)-induced hepatic steatosis. EF-2001 treatment inhibited the oleic acid (OA) lipid accumulation in FL83B liver cells. Furthermore, we performed lipid reduction analysis to confirm the underlying mechanism of lipolysis. The results showed that EF-2001 downregulated proteins and upregulated AMP-activated protein kinase (AMPK) phosphorylation in the sterol regulatory element-binding protein 1c (SREBP-1c) and AMPK signaling pathways, respectively. The effect of EF-2001 on OA-induced hepatic lipid accumulation in FL83Bs enhanced the phosphorylation of acetyl-CoA carboxylase and reduced the levels of lipid accumulation proteins SREBP-1c and fatty acid synthase. EF-2001 treatment increased the levels of adipose triglyceride lipase and monoacylglycerol during lipase enzyme activation, which, when increased, contributed to increased liver lipolysis. In conclusion, EF-2001 inhibits OA-induced FL83B hepatic lipid accumulation and HFD-induced hepatic steatosis in rats through the AMPK signaling pathway.
Collapse
Affiliation(s)
- Jin-Ho Lee
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Keun-Jung Woo
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Joonpyo Hong
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Kwon-Il Han
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
- Research & Development Center, Bereum Co., Ltd., Wonju 26361, Republic of Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Tack-Joong Kim
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
- Research & Development Center, Doctor TJ Co., Ltd., Wonju 26493, Republic of Korea
- Correspondence: ; Tel.: +82-33-760-224
| |
Collapse
|
13
|
Tonelli A, Lumngwena EN, Ntusi NAB. The oral microbiome in the pathophysiology of cardiovascular disease. Nat Rev Cardiol 2023; 20:386-403. [PMID: 36624275 DOI: 10.1038/s41569-022-00825-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
Despite advances in our understanding of the pathophysiology of many cardiovascular diseases (CVDs) and expansion of available therapies, the global burden of CVD-associated morbidity and mortality remains unacceptably high. Important gaps remain in our understanding of the mechanisms of CVD and determinants of disease progression. In the past decade, much research has been conducted on the human microbiome and its potential role in modulating CVD. With the advent of high-throughput technologies and multiomics analyses, the complex and dynamic relationship between the microbiota, their 'theatre of activity' and the host is gradually being elucidated. The relationship between the gut microbiome and CVD is well established. Much less is known about the role of disruption (dysbiosis) of the oral microbiome; however, interest in the field is growing, as is the body of literature from basic science and animal and human investigations. In this Review, we examine the link between the oral microbiome and CVD, specifically coronary artery disease, stroke, peripheral artery disease, heart failure, infective endocarditis and rheumatic heart disease. We discuss the various mechanisms by which oral dysbiosis contributes to CVD pathogenesis and potential strategies for prevention and treatment.
Collapse
Affiliation(s)
- Andrea Tonelli
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cardiovascular Research Unit, Christiaan Barnard Division of Cardiothoracic Surgery, Department of Surgery, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa
| | - Evelyn N Lumngwena
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.,Centre for the Study of Emerging and Re-emerging Infections, Institute for Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa. .,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa. .,Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Wellcome Centre for Infectious Disease Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
14
|
Zhao N, Wang Y, Ma Y, Liang X, Zhang X, Gao Y, Dong Y, Bai D, Hu J. Jia-Wei-Si-Miao-Yong-An decoction modulates intestinal flora and metabolites in acute coronary syndrome model. Front Cardiovasc Med 2023; 9:1038273. [PMID: 36684592 PMCID: PMC9845626 DOI: 10.3389/fcvm.2022.1038273] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Aims We assessed the efficacy of the traditional Chinese medicine formulation Jia-Wei-Si-Miao-Yong-An decoction (HJ11) in the treatment of acute coronary syndrome and evaluated its impact on the intestinal microbiota and their metabolites. Methods An acute coronary syndrome model was established in rats, which were randomly assigned to the model, HJ11 treatment, and atorvastatin treatment groups. Rats were then administered saline solution (model and sham operation control groups) or drugs by oral gavage for 28 d. Echocardiography was performed and serum creatine kinase-MB and cardiac troponin I levels were monitored to examine the cardiac function. Inflammation was evaluated using hematoxylin and eosin staining of heart tissue, and serum interleukin-2, interleukin-6, tumor necrosis factor alpha, and high-sensitivity C-reactive protein measurements. Gut microbiota composition was analyzed via 16S rRNA gene sequencing. Metabolomics was used to determine fecal metabolites and elucidate the modes of action of HJ11 in acute coronary syndrome treatment. Results HJ11 improved cardiac function and attenuated inflammation in rats with acute coronary syndrome. Relative to the untreated model group, the HJ11-treated group presented normalized Firmicutes/Bacteroidetes ratio and reduced abundances of the bacterial genera norank_f__Ruminococcaceae, Desulfovibrio, Clostridium_sensu_stricto_1, Adlercreutzia, Staphylococcus, Bacteroides, Prevotella, Rikenellaceae_RC9_gut_group, unclassified_o__Bacteroidales, and Ruminococcus_gauvreauii_group. We found 23 differentially expressed intestinal metabolites, and the enriched metabolic pathways were mainly related to amino acid metabolism. We also discovered that asymmetric dimethylarginine levels were strongly associated with cardiovascular disease. Correlation analyses revealed strong associations among intestinal microflora, their metabolites, proinflammatory factors, and cardiac function. Hence, the therapeutic effects of HJ11 on acute coronary syndrome are related to specific alterations in gut microbiota and their metabolites. Conclusion This work demonstrated that HJ11 effectively treats acute coronary syndrome. HJ11 seems to increase the abundance of beneficial bacterial taxa (Bacteroides and Rikenellaceae_RC9_gut_group), mitigate the risk factors associated with cardiovascular disease, alter bacterial metabolites, lower asymmetric dimethylarginine levels, and effectively treat acute coronary syndrome.
Collapse
Affiliation(s)
- Ning Zhao
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Pharmacy, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Wang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Ma
- Department of Pathophysiology and Allergy Research, Vienna General Hospital, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Xiaoxue Liang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xi Zhang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuan Gao
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Dong
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dong Bai
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingqing Hu
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Fan Y, Liang L, Tang X, Zhu J, Mu L, Wang M, Huang X, Gong S, Xu J, Liu T, Zhang T. Changes in the gut microbiota structure and function in rats with doxorubicin-induced heart failure. Front Cell Infect Microbiol 2023; 13:1135428. [PMID: 37180435 PMCID: PMC10173310 DOI: 10.3389/fcimb.2023.1135428] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Objectives The rat model of heart failure (HF) induced by doxorubicin (DOX), a broad spectrum and highly effective chemotherapeutic anthracycline with high-affinity to myocardial tissue that causes severe dose-dependent irreversible cardiotoxicity has been widely recognized and applied in HF pathogenesis and drug therapy studies. The gut microbiota (GM) has attracted significant attention due to its potential role in HF, and research in this area may provide beneficial therapeutic strategies for HF. Considering the differences in the route, mode, and total cumulative dose of DOX administration used to establish HF models, the optimal scheme for studying the correlation between GM and HF pathogenesis remains to be determined. Therefore, focusing on establishing the optimal scheme, we evaluated the correlation between GM composition/function and DOX-induced cardiotoxicity (DIC). Methods Three schemes were investigated: DOX (at total cumulative doses of 12, 15 or 18 mg/kg using a fixed or alternating dose via a tail vein or intraperitoneal injection) was administered to Sprague Dawley (SD) for six consecutive weeks. The M-mode echocardiograms performed cardiac function evaluation. Pathological changes in the intestine were observed by H&E staining and in the heart by Masson staining. The serum levels of N-terminal pre-B-type natriuretic peptide (NT-proBNP) and cardiac troponin I (cTnI) were measured by ELISA. The GM was analysed by 16S rRNA gene sequencing. Key findings Strikingly, based on the severity of cardiac dysfunction, there were marked differences in the abundance and grouping of GM under different schemes. The HF model established by tail vein injection of DOX (18 mg/kg, alternating doses) was more stable; moreover, the degree of myocardial injury and microbial composition were more consistent with the clinical manifestations of HF. Conclusions The model of HF established by tail vein injection of doxorubicin, administered at 4mg/kg body weight (2mL/kg) at weeks 1, 3 and 5, and at 2mg/kg body weight (1mL/kg) at weeks 2, 4 and 6, with a cumulative total dose of 18mg/kg, is a better protocol to study the correlation between HF and GM.
Collapse
Affiliation(s)
- Yawen Fan
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Lichang Liang
- Department of Preventive Treatment, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xinzheng Tang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jinxian Zhu
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Lei Mu
- Department of Encephalopathy Diseases, Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen, China
| | - Mengni Wang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xuecheng Huang
- Department of Spinal Surgery, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Shenglan Gong
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Jinghan Xu
- Department of Endocrinology, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Tianjiao Liu
- Department of Endocrinology, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Tianfeng Zhang
- Department of Cardiovascular Diseases, The Sixth Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
- *Correspondence: Tianfeng Zhang,
| |
Collapse
|
16
|
Salazar J, Morillo V, Suárez MK, Castro A, Ramírez P, Rojas M, Añez R, D’Marco L, Chacín-González M, Bermudez V. Role of Gut Microbiome in Atherosclerosis: Molecular and Therapeutic Aspects. Curr Cardiol Rev 2023; 19:e020223213408. [PMID: 36733248 PMCID: PMC10494273 DOI: 10.2174/1573403x19666230202164524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is one of the most relevant and prevalent cardiovascular diseases of our time. It is one of the pathological entities that increases the morbidity and mortality index in the adult population. Pathophysiological connections have been observed between atherosclerosis and the gut microbiome (GM), represented by a group of microorganisms that are present in the gut. These microorganisms are vital for metabolic homeostasis in humans. Recently, direct and indirect mechanisms through which GM can affect the development of atherosclerosis have been studied. This has led to research into the possible modulation of GM and metabolites as a new target in the prevention and treatment of atherosclerosis. The goal of this review is to analyze the physiopathological mechanisms linking GM and atherosclerosis that have been described so far. We also aim to summarize the recent studies that propose GM as a potential target in atherosclerosis management.
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Valery Morillo
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María K Suárez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ana Castro
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Paola Ramírez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición. Hospital General Universitario Gregorio Marañón, Madrid, España
| | - Luis D’Marco
- Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, 46115, Spain
| | | | - Valmore Bermudez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| |
Collapse
|
17
|
Pavlidou E, Fasoulas A, Mantzorou M, Giaginis C. Clinical Evidence on the Potential Beneficial Effects of Probiotics and Prebiotics in Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms232415898. [PMID: 36555535 PMCID: PMC9779729 DOI: 10.3390/ijms232415898] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
The 'gut microbiome'-the hundreds of trillions of bacteria in the human gastrointestinal tract-serves several functions. The gut microbiome includes all the microorganisms, bacteria, viruses, protozoa, and fungi in the gastrointestinal tract and their genetic material. It helps digest indigestible foods and produces nutrients. Through the metabolism of sugars and proteins, it helps the intestinal barrier, the immune system, and metabolism. Some bacteria, such as those in the gut microbiome, cause disease, but others are essential to our health. These "good" microbes protect us from pathogens. Numerous studies have linked an unhealthy gut microbiome to obesity, insulin resistance, depression, and cardiometabolic risk factors. To maximize probiotic benefits in each case, knowledge of probiotic bacterial strains and how to consume them should be increased. This study aims to examine the benefits of probiotic and prebiotic organisms on cardiovascular health, specifically on heart disease, coronary heart disease, stroke, and hypertension. To complete the research, a literature review was conducted by gathering clinical studies and data. The clinical evidence demonstrates the beneficial effect of probiotics and prebiotic microorganisms on the gut microbiome, which has multiple benefits for overall health and especially for cardiovascular diseases.
Collapse
|
18
|
Fu Z, Song X, Shen A, Zhou T. Microarray analysis reveals the potential molecular mechanism of Lp299v in stable coronary atherosclerotic disease. AMB Express 2022; 12:125. [PMID: 36152115 PMCID: PMC9509519 DOI: 10.1186/s13568-022-01466-y] [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: 07/06/2022] [Accepted: 09/11/2022] [Indexed: 11/17/2022] Open
Abstract
A growing body of evidence has confirmed that inflammatory mechanisms are involved in the formation and treatment of coronary atherosclerotic disease (CAD). An increase in circulatory levels of inflammatory cytokines has been found in patients with CAD, while the molecular mechanisms of inflammation still remain elusive. This study was designed to identify differentially expressed genes (DEGs), and to explore the molecular mechanism and hub genes that are involved in the effects of Lactobacillus plantarum 299v (Lp299v) supplementation. Microarray dataset (GSE156357) was downloaded from the Gene Expression Omnibus (GEO) database. The DEGs were identified by the R software. Then, the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses and construction of protein–protein interaction (PPI) network were performed by DAVID, STRING, and Cytoscape software. In daily alcohol user (DAU) group, 7,541 DEGs were identified, including 206 up-regulated and 7,335 down-regulated DEGs. In non-daily alcohol user (non-DAU) group, 2,799 DEGs were identified (2,491 up-regulated and 308 down-regulated DEGs). The GO enrichment analysis revealed that miosis was up-regulated and immune response was down-regulated. The KEGG enrichment analysis showed that Lp299v supplementation reduced the levels of chemotactic cytokines, and weakened immune response. Proteins of G protein-coupled receptor, inflammatory response, regulation of cell proliferation and apoptosis-related proteins were found in the PPI network. The hub genes were associated with G protein-coupled receptor, inflammatory response, and cell proliferation and apoptosis. The weighted gene co-expression network analysis (WGCNA) enriched the DEGs in 4 modules. This study indicated the expressions of chemokine receptors and regulation of immune response in the Lp299v supplementation. Meanwhile, it was supposed that chemokine receptors may have a cellular effect.
Collapse
Affiliation(s)
- Zhenyang Fu
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510630, China
| | - Xiaolei Song
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University,Guangzhou Medical University, Guangzhou, China
| | - Anna Shen
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510630, China
| | - Tao Zhou
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
19
|
Mutalub YB, Abdulwahab M, Mohammed A, Yahkub AM, AL-Mhanna SB, Yusof W, Tang SP, Rasool AHG, Mokhtar SS. Gut Microbiota Modulation as a Novel Therapeutic Strategy in Cardiometabolic Diseases. Foods 2022; 11:2575. [PMID: 36076760 PMCID: PMC9455664 DOI: 10.3390/foods11172575] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
The human gut harbors microbial ecology that is in a symbiotic relationship with its host and has a vital function in keeping host homeostasis. Inimical alterations in the composition of gut microbiota, known as gut dysbiosis, have been associated with cardiometabolic diseases. Studies have revealed the variation in gut microbiota composition in healthy individuals as compared to the composition of those with cardiometabolic diseases. Perturbation of host-microbial interaction attenuates physiological processes and may incite several cardiometabolic disease pathways. This imbalance contributes to cardiometabolic diseases via metabolism-independent and metabolite-dependent pathways. The aim of this review was to elucidate studies that have demonstrated the complex relationship between the intestinal microbiota as well as their metabolites and the development/progression of cardiometabolic diseases. Furthermore, we systematically itemized the potential therapeutic approaches for cardiometabolic diseases that target gut microbiota and/or their metabolites by following the pathophysiological pathways of disease development. These approaches include the use of diet, prebiotics, and probiotics. With the exposition of the link between gut microbiota and cardiometabolic diseases, the human gut microbiota therefore becomes a potential therapeutic target in the development of novel cardiometabolic agents.
Collapse
Affiliation(s)
- Yahkub Babatunde Mutalub
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia or
- Department of Clinical Pharmacology, College of Medical Sciences, Abubakar Tafawa Balewa University, Bauchi 74027, Nigeria
| | - Monsurat Abdulwahab
- Department of Midwifery, College of Nursing Sciences, Abubakar Tafawa Balewa University Teaching Hospital, Bauchi 74027, Nigeria
| | - Alkali Mohammed
- Department of Medicine, College of Medical Sciences, Abubakar Tafawa Balewa University, Bauchi 74027, Nigeria
| | - Aishat Mutalib Yahkub
- College of Medical Sciences, Abubakar Tafawa Balewa University, Bauchi 74027, Nigeria
| | - Sameer Badri AL-Mhanna
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Wardah Yusof
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Suk Peng Tang
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia or
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia or
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia or
| |
Collapse
|
20
|
Luo Y, Zhang Y, Han X, Yuan Y, Zhou Y, Gao Y, Yu H, Zhang J, Shi Y, Duan Y, Zhao X, Yan S, Hao H, Dai C, Zhao S, Shi J, Li W, Zhang S, Xu W, Fang N, Gong Y, Li Y. Akkermansia muciniphila prevents cold-related atrial fibrillation in rats by modulation of TMAO induced cardiac pyroptosis. EBioMedicine 2022; 82:104087. [PMID: 35797768 PMCID: PMC9270211 DOI: 10.1016/j.ebiom.2022.104087] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Background Cold exposure is one of the most important risk factors for atrial fibrillation (AF), and closely related to the poor prognosis of AF patients. However, the mechanisms underlying cold-related AF are poorly understood. Methods Various techniques including 16S rRNA gene sequencing, fecal microbiota transplantation, and electrophysiological examination were used to determine whether gut microbiota dysbiosis promotes cold-related AF. Metabonomics were performed to investigate changes in fecal trimethylamine (TMA) and plasma trimethylamine N-oxide (TMAO) during cold exposure. The detailed mechanism underlying cold-related AF were examined in vitro. Transgenic mice were constructed to explore the role of pyroptosis in cold-related AF. The human cohort was used to evaluate the correlation between A. muciniphila and cold-related AF. Findings We found that cold exposure caused elevated susceptibility to AF and reduced abundance of Akkermansia muciniphila (A. muciniphila) in rats. Intriguingly, oral supplementation of A. muciniphila ameliorated the pro-AF property induced by cold exposure. Mechanistically, cold exposure disrupted the A. muciniphila, by which elevated the level of trimethylamine N-oxide (TMAO) through modulation of the microbial enzymes involved in trimethylamine (TMA) synthesis. Correspondingly, progressively increased plasma TMAO levels were validated in human subjects during cold weather. Raised TMAO enhanced the infiltration of M1 macrophages in atria and increased the expression of Casp1-p20 and cleaved-GSDMD, ultimately causing atrial structural remodeling. Furthermore, the mice with conditional deletion of caspase1 exhibited resistance to cold-related AF. More importantly, a cross-sectional clinical study revealed that the reduction of A. muciniphila abundance was an independent risk factor for cold-related AF in human subjects. Interpretation Our findings revealed a novel causal role of aberrant gut microbiota and metabolites in pathogenesis of cold-related AF, which raises the possibility of selectively targeting microbiota and microbial metabolites as a potential therapeutic strategy for cold-related AF. Funding This work was supported by grants from the State Key Program of National Natural Science Foundation of China (No.81830012), and National Natural Science Foundation of China (No.82070336, No.81974024), Youth Program of the National Natural Science Foundation of China (No.81900374, No.81900302), and Excellent Young Medical Talents supporting project in the First Affiliated Hospital of Harbin Medical University (No. HYD2020YQ0001).
Collapse
Affiliation(s)
- Yingchun Luo
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yun Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China; Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuejie Han
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yue Yuan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yun Zhou
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yunlong Gao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Hui Yu
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Jiawei Zhang
- Department of Cardiology, Qingdao Central Hospital, Qingdao, China
| | - Yiya Shi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Duan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Xinbo Zhao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Sen Yan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Hongting Hao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Chenguang Dai
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Shiqi Zhao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Jing Shi
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Wenpeng Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Song Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Wei Xu
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Ning Fang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yongtai Gong
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China.
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China; NHC Key Laboratory of Cell Translation, Harbin Medical University, Heilongjiang 150001, China; Key Laboratory of Hepatosplenic Surgery, Harbin Medical University, Ministry of Education, Harbin 150001, China; Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin 150001, China; Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Harbin 150081, China.
| |
Collapse
|
21
|
The heart and gut relationship: a systematic review of the evaluation of the microbiome and trimethylamine-N-oxide (TMAO) in heart failure. Heart Fail Rev 2022; 27:2223-2249. [PMID: 35726110 DOI: 10.1007/s10741-022-10254-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 02/08/2023]
Abstract
There is an expanding body of research on the bidirectional relationship of the human gut microbiome and cardiovascular disease, including heart failure (HF). Researchers are examining the microbiome and gut metabolites, primarily trimethylamine-N-oxide (TMAO), to understand clinically observed outcomes. This systematic review explored the current state of the science on the evaluation and testing of the gut biome in persons with HF. Using electronic search methods of Medline, Embase, CINAHL, and Web of Science, until December 2021, we identified 511 HF biome investigations between 2014 and 2021. Of the 30 studies included in the review, six were 16S rRNA and nineteen TMAO, and three both TMAO and 16S rRNA, and two bacterial cultures. A limited range of study designs were represented, the majority involving single cohorts (n = 10) and comparing individuals with HF to controls (n = 15). Patients with HF had less biodiversity in fecal samples compared to controls. TMAO is associated with age, BNP, eGFR, HF severity, and poor outcomes including hospitalizations and mortality. Inconsistent across studies was the ability of TMAO to predict HF development, the independent prognostic value of TMAO when controlling for renal indices, and the relationship of TMAO to LVEF and CRP. Gut microbiome dysbiosis is associated with HF diagnosis, disease severity, and prognostication related to hospitalizations and mortality. Gut microbiome research in patients with HF is developing. Further longitudinal and multi-centered studies are required to inform interventions to promote clinical decision-making and improved patient outcomes.
Collapse
|
22
|
Shulpekova Y, Zharkova M, Tkachenko P, Tikhonov I, Stepanov A, Synitsyna A, Izotov A, Butkova T, Shulpekova N, Lapina N, Nechaev V, Kardasheva S, Okhlobystin A, Ivashkin V. The Role of Bile Acids in the Human Body and in the Development of Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113401. [PMID: 35684337 PMCID: PMC9182388 DOI: 10.3390/molecules27113401] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/13/2022] [Accepted: 05/23/2022] [Indexed: 11/28/2022]
Abstract
Bile acids are specific and quantitatively important organic components of bile, which are synthesized by hepatocytes from cholesterol and are involved in the osmotic process that ensures the outflow of bile. Bile acids include many varieties of amphipathic acid steroids. These are molecules that play a major role in the digestion of fats and the intestinal absorption of hydrophobic compounds and are also involved in the regulation of many functions of the liver, cholangiocytes, and extrahepatic tissues, acting essentially as hormones. The biological effects are realized through variable membrane or nuclear receptors. Hepatic synthesis, intestinal modifications, intestinal peristalsis and permeability, and receptor activity can affect the quantitative and qualitative bile acids composition significantly leading to extrahepatic pathologies. The complexity of bile acids receptors and the effects of cross-activations makes interpretation of the results of the studies rather difficult. In spite, this is a very perspective direction for pharmacology.
Collapse
Affiliation(s)
- Yulia Shulpekova
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Maria Zharkova
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Pyotr Tkachenko
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Igor Tikhonov
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Alexander Stepanov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | - Alexandra Synitsyna
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
- Correspondence: ; Tel.: +7-499-764-98-78
| | - Alexander Izotov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | - Tatyana Butkova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119435 Moscow, Russia; (A.S.); (A.I.); (T.B.)
| | | | - Natalia Lapina
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Vladimir Nechaev
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Svetlana Kardasheva
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Alexey Okhlobystin
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| | - Vladimir Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; (Y.S.); (M.Z.); (P.T.); (I.T.); (N.L.); (V.N.); (S.K.); (A.O.); (V.I.)
| |
Collapse
|
23
|
Sumida K, Han Z, Chiu CY, Mims TS, Bajwa A, Demmer RT, Datta S, Kovesdy CP, Pierre JF. Circulating Microbiota in Cardiometabolic Disease. Front Cell Infect Microbiol 2022; 12:892232. [PMID: 35592652 PMCID: PMC9110890 DOI: 10.3389/fcimb.2022.892232] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
The rapid expansion of microbiota research has significantly advanced our understanding of the complex interactions between gut microbiota and cardiovascular, metabolic, and renal system regulation. Low-grade chronic inflammation has long been implicated as one of the key mechanisms underlying cardiometabolic disease risk and progression, even before the insights provided by gut microbiota research in the past decade. Microbial translocation into the bloodstream can occur via different routes, including through the oral and/or intestinal mucosa, and may contribute to chronic inflammation in cardiometabolic disease. Among several gut-derived products identifiable in the systemic circulation, bacterial endotoxins and metabolites have been extensively studied, however recent advances in microbial DNA sequencing have further allowed us to identify highly diverse communities of microorganisms in the bloodstream from an -omics standpoint, which is termed "circulating microbiota." While detecting microorganisms in the bloodstream was historically considered as an indication of infection, evidence on the circulating microbiota is continually accumulating in various patient populations without clinical signs of infection and even in otherwise healthy individuals. Moreover, both quantitative and compositional alterations of the circulating microbiota have recently been implicated in the pathogenesis of chronic inflammatory conditions, potentially through their immunostimulatory, atherogenic, and cardiotoxic properties. In this mini review, we aim to provide recent evidence on the characteristics and roles of circulating microbiota in several cardiometabolic diseases, such as type 2 diabetes, cardiovascular disease, and chronic kidney disease, with highlights of our emerging findings on circulating microbiota in patients with end-stage kidney disease undergoing hemodialysis.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,*Correspondence: Keiichi Sumida,
| | - Zhongji Han
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chi-Yang Chiu
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Tahliyah S. Mims
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Amandeep Bajwa
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Csaba P. Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,Nephrology Section, Memphis Veterans Affairs (VA) Medical Center, Memphis, TN, United States
| | - Joseph F. Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
24
|
Gut–Skin Axis: Unravelling the Connection between the Gut Microbiome and Psoriasis. Biomedicines 2022; 10:biomedicines10051037. [PMID: 35625774 PMCID: PMC9138548 DOI: 10.3390/biomedicines10051037] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
Evidence has shown that gut microbiome plays a role in modulating the development of diseases beyond the gastrointestinal tract, including skin disorders such as psoriasis. The gut–skin axis refers to the bidirectional relationship between the gut microbiome and skin health. This is regulated through several mechanisms such as inflammatory mediators and the immune system. Dysregulation of microbiota has been seen in numerous inflammatory skin conditions such as atopic dermatitis, rosacea, and psoriasis. Understanding how gut microbiome are involved in regulating skin health may lead to development of novel therapies for these skin disorders through microbiome modulation, in particularly psoriasis. In this review, we will compare the microbiota between psoriasis patients and healthy control, explain the concept of gut–skin axis and the effects of gut dysbiosis on skin physiology. We will also review the current evidence on modulating gut microbiome using probiotics in psoriasis.
Collapse
|
25
|
Henneke L, Schlicht K, Andreani NA, Hollstein T, Demetrowitsch T, Knappe C, Hartmann K, Jensen-Kroll J, Rohmann N, Pohlschneider D, Geisler C, Schulte DM, Settgast U, Türk K, Zimmermann J, Kaleta C, Baines JF, Shearer J, Shah S, Shen-Tu G, Schwarz K, Franke A, Schreiber S, Laudes M. A dietary carbohydrate - gut Parasutterella - human fatty acid biosynthesis metabolic axis in obesity and type 2 diabetes. Gut Microbes 2022; 14:2057778. [PMID: 35435797 PMCID: PMC9037427 DOI: 10.1080/19490976.2022.2057778] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent rodent microbiome experiments suggest that besides Akkermansia, Parasutterella sp. are important in type 2 diabetes and obesity development. In the present translational human study, we aimed to characterize Parasutterella in our European cross-sectional FoCus cohort (n = 1,544) followed by validation of the major results in an independent Canadian cohort (n = 438). In addition, we examined Parasutterella abundance in response to a weight loss intervention (n = 55). Parasutterella was positively associated with BMI and type 2 diabetes independently of the reduced microbiome α/β diversity and low-grade inflammation commonly found in obesity. Nutritional analysis revealed a positive association with the dietary intake of carbohydrates but not with fat or protein consumption. Out of 126 serum metabolites differentially detectable by untargeted HPLC-based MS-metabolomics, L-cysteine showed the strongest reduction in subjects with high Parasutterella abundance. This is of interest, since Parasutterella is a known high L-cysteine consumer and L-cysteine is known to improve blood glucose levels in rodents. Furthermore, metabolic network enrichment analysis identified an association of high Parasutterella abundance with the activation of the human fatty acid biosynthesis pathway suggesting a mechanism for body weight gain. This is supported by a significant reduction of the Parasutterella abundance during our weight loss intervention. Together, these data indicate a role for Parasutterella in human type 2 diabetes and obesity, whereby the link to L-cysteine might be relevant in type 2 diabetes development and the link to the fatty acid biosynthesis pathway for body weight gain in response to a carbohydrate-rich diet in obesity development.
Collapse
Affiliation(s)
- Lea Henneke
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Kristina Schlicht
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Nadia A. Andreani
- Section of Evolutionary Medicine, Institute for Experimental Medicine University of Kiel, Kiel, Germany,Guest group for evolutionary medicine Max-Planck-Institute of Evolutionary Biology, Plön, Germany
| | - Tim Hollstein
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Tobias Demetrowitsch
- Division of Food Technology, Department of Human Nutrition, University of Kiel, Kiel, Germany
| | - Carina Knappe
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Katharina Hartmann
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Julia Jensen-Kroll
- Division of Food Technology, Department of Human Nutrition, University of Kiel, Kiel, Germany
| | - Nathalie Rohmann
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Daniela Pohlschneider
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Corinna Geisler
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Dominik M. Schulte
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Ute Settgast
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Kathrin Türk
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany
| | - Johannes Zimmermann
- Research Group Medical System Biology, Institute of Experimental Medicine, University of Kiel, Kiel, Germany
| | - Christoph Kaleta
- Research Group Medical System Biology, Institute of Experimental Medicine, University of Kiel, Kiel, Germany
| | - John F. Baines
- Section of Evolutionary Medicine, Institute for Experimental Medicine University of Kiel, Kiel, Germany,Guest group for evolutionary medicine Max-Planck-Institute of Evolutionary Biology, Plön, Germany
| | - Jane Shearer
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Faculty Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Shrushti Shah
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Faculty Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Grace Shen-Tu
- Alberta’s Tomorrow Project, Cancer Control Alberta, Alberta Health Services, Edmonton, AB, Canada
| | - Karin Schwarz
- Division of Food Technology, Department of Human Nutrition, University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Matthias Laudes
- Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Kiel, Germany,Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Medicine 1, University Medical Centre Schleswig-Holstein, Kiel University, Kiel, Germany,CONTACT Matthias Laudes Institute of Diabetes and Clinical Metabolic Research, University of Kiel, Düsternbrooker Weg 17, 24105Kiel, Germany
| |
Collapse
|
26
|
Clinical Phenotypes of Cardiovascular and Heart Failure Diseases Can Be Reversed? The Holistic Principle of Systems Biology in Multifaceted Heart Diseases. CARDIOGENETICS 2022. [DOI: 10.3390/cardiogenetics12020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Recent advances in cardiology and biological sciences have improved quality of life in patients with complex cardiovascular diseases (CVDs) or heart failure (HF). Regardless of medical progress, complex cardiac diseases continue to have a prolonged clinical course with high morbidity and mortality. Interventional coronary techniques together with drug therapy improve quality and future prospects of life, but do not reverse the course of the atherosclerotic process that remains relentlessly progressive. The probability of CVDs and HF phenotypes to reverse can be supported by the advances made on the medical holistic principle of systems biology (SB) and on artificial intelligence (AI). Studies on clinical phenotypes reversal should be based on the research performed in large populations of patients following gathering and analyzing large amounts of relative data that embrace the concept of complexity. To decipher the complexity conundrum, a multiomics approach is needed with network analysis of the biological data. Only by understanding the complexity of chronic heart diseases and explaining the interrelationship between different interconnected biological networks can the probability for clinical phenotypes reversal be increased.
Collapse
|
27
|
Liao W, Khan I, Huang G, Chen S, Liu L, Leong WK, Li XA, Wu J, Wendy Hsiao WL. Bifidobacterium animalis: the missing link for the cancer-preventive effect of Gynostemma pentaphyllum. Gut Microbes 2022; 13:1847629. [PMID: 33228450 PMCID: PMC8381792 DOI: 10.1080/19490976.2020.1847629] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) ranks the third most common cancer type in both men and women. Besides the known genetic and epigenetic changes in the gut epithelial cells, we now know that disturbed gut microbes could also contribute to the onset and progression of CRC. Hence, keeping a balanced gut microbiota (GM) has become a novel pursue in the medical field, particularly in the area of gastrointestinal disorders. Gynostemma pentaphyllum (Gp) is a dietary herbal medicine. In our previous study, Gp saponins (GpS) displayed prebiotic and cancer-preventive properties through the modulation of GM in ApcMin/+ mice. However, the specific group(s) of GM links to the health effects of GpS remains unknown. To track down the missing link, we first investigated and found that inoculation with fecal materials from GpS-treated ApcMin/+ mice effectively reduces polyps in ApcMin/+ mice. From the same source of the fecal sample, we successfully isolated 16 bacterial species. Out of the 16 bacteria, Bifidobacterium animalis stands out as the responder to the GpS-growth stimulus. Biochemical and RNAseq analysis demonstrated that GpS enhanced expressions of a wide range of genes encoding biogenesis and metabolic pathways in B. animalis culture. Moreover, we found that colonization of B. animalis markedly reduces the polyp burden in ApcMin/+ mice. These findings reveal a mutualistic interaction between the prebiotic and a probiotic to achieve anticancer and cancer-preventive activities. Our result, for the first time, unveils the anticancer function of B. animalis and extend the probiotic horizon of B. animalis.
Collapse
Affiliation(s)
- Weilin Liao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Guoxin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Shengshuang Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Wai Kit Leong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Xiao Ang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - Jianlin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| | - W. L. Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR,CONTACT W. L. Wendy Hsiao State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR
| |
Collapse
|
28
|
Fang C, Zuo K, Zhang W, Zhong J, Li J, Xu L, Yang X. Association between Gut Microbiota Dysbiosis and the CHA2DS2-VASc Score in Atrial Fibrillation Patients. Int J Clin Pract 2022; 2022:7942605. [PMID: 35685549 PMCID: PMC9159190 DOI: 10.1155/2022/7942605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In our previous studies, we found a disordered taxonomic composition and function of gut microbiota (GM) in atrial fibrillation (AF) patients. However, direct evidence about the association between dysbiotic microbiota and thromboembolic risk in AF is lacking. AIMS In this study, we analyzed the interaction of GM and related functional patterns in AF with different CHA2DS2-VASc scores to assess its potential as a biomarker for predicting stroke risk. Patients and Methods. The CHA2DS2-VASc score was used for thromboembolic risk stratification in AF according to American Heart Association (AHA) guidelines. We investigated the taxonomic and functional annotation of GM based on metagenomic data from 50 AF patients (32 with high thromboembolic risk (CHA2DS2-VASc score ≥2 (males) or CHA2DS2-VASc score ≥3 (females)) and 18 individuals with low thromboembolic risk (CHA2DS2-VASc score <2 (males) or CHA2DS2-VASc score <3 (females))). RESULTS The gut microbial diversity, composition, and function in AF were different in high and low CHA2DS2-VASc score groups. In high thromboembolic risk group, the abundance of Prevotella, Lachnospiraceae, and Eubacterium rectale, related to the production of short-chain fatty acids and anti-inflammatory were reduced (all P < 0.05). Furthermore, annotated by Kyoto Encyclopedia of Genes and Genomes (KEGG), a database of genes and genomes, the KEGG orthology-based scoring approach exhibited a significant association with thromboembolic risk in AF patients. CONCLUSIONS Imbalance of GM and microbial dysfunction are involved in aggravated thromboembolic risk of AF.
Collapse
Affiliation(s)
- Chen Fang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Kun Zuo
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wanjing Zhang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiuchang Zhong
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jing Li
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Li Xu
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xinchun Yang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
29
|
Lad N, Murphy A, Parenti C, Nelson C, Williams N, Sharpe G, McTernan P. Asthma and obesity: endotoxin another insult to add to injury? Clin Sci (Lond) 2021; 135:2729-2748. [PMID: 34918742 PMCID: PMC8689194 DOI: 10.1042/cs20210790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Low-grade inflammation is often an underlying cause of several chronic diseases such as asthma, obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Defining the mediators of such chronic low-grade inflammation often appears dependent on which disease is being investigated. However, downstream systemic inflammatory cytokine responses in these diseases often overlap, noting there is no doubt more than one factor at play to heighten the inflammatory response. Furthermore, it is increasingly believed that diet and an altered gut microbiota may play an important role in the pathology of such diverse diseases. More specifically, the inflammatory mediator endotoxin, which is a complex lipopolysaccharide (LPS) derived from the outer membrane cell wall of Gram-negative bacteria and is abundant within the gut microbiota, and may play a direct role alongside inhaled allergens in eliciting an inflammatory response in asthma. Endotoxin has immunogenic effects and is sufficiently microscopic to traverse the gut mucosa and enter the systemic circulation to act as a mediator of chronic low-grade inflammation in disease. Whilst the role of endotoxin has been considered in conditions of obesity, cardiovascular disease and T2DM, endotoxin as an inflammatory trigger in asthma is less well understood. This review has sought to examine the current evidence for the role of endotoxin in asthma, and whether the gut microbiota could be a dietary target to improve disease management. This may expand our understanding of endotoxin as a mediator of further low-grade inflammatory diseases, and how endotoxin may represent yet another insult to add to injury.
Collapse
Affiliation(s)
- Nikita Lad
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Alice M. Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Cristina Parenti
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Carl P. Nelson
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Neil C. Williams
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Graham R. Sharpe
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Philip G. McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| |
Collapse
|
30
|
Tuerhongjiang G, Guo M, Qiao X, Lou B, Wang C, Wu H, Wu Y, Yuan Z, She J. Interplay Between Gut Microbiota and Amino Acid Metabolism in Heart Failure. Front Cardiovasc Med 2021; 8:752241. [PMID: 34746265 PMCID: PMC8566708 DOI: 10.3389/fcvm.2021.752241] [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: 08/02/2021] [Accepted: 09/07/2021] [Indexed: 11/14/2022] Open
Abstract
Heart failure (HF) is a complex clinical syndrome of which the incidence is on the rise worldwide. Cardiometabolic disorders are associated with the deterioration of cardiac function and progression of HF. Recently, there has been renewed interest in gut microbiota (GM) and its metabolites in the cardiovascular disease. HF-caused hypoperfusion could increase intestinal permeability, and a “leaky” bowel leads to bacterial translocation and make its metabolites more easily enter the circulation. Considerable evidence shows that the composition of microbiota and amino acids (AAs) has been altered in HF patients, and AAs could serve as a diagnostic and prognostic biomarker in HF. The findings indicate that the gut–amino acid–HF axis may play a key role in the progression of HF. In this paper, we focus on the interrelationship between the AA metabolism and GM alterations during the development of heart failure. We also discuss the potential prognostic and therapeutic value of the gut–amino acid–HF axis in the cortex of HF.
Collapse
Affiliation(s)
- Gulinigaer Tuerhongjiang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Manyun Guo
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Xiangrui Qiao
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Bowen Lou
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Chen Wang
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Haoyu Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Yue Wu
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Zuyi Yuan
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| | - Jianqing She
- Department of Cardiovascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, China
| |
Collapse
|
31
|
Gut Microbiome, Functional Food, Atherosclerosis, and Vascular Calcifications-Is There a Missing Link? Microorganisms 2021; 9:microorganisms9091913. [PMID: 34576810 PMCID: PMC8472650 DOI: 10.3390/microorganisms9091913] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/21/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome is represented by the genome of all microorganisms (symbiotic, potential pathogens, or pathogens) residing in the intestine. These ecological communities are involved in almost all metabolic diseases and cardiovascular diseases are not excluded. Atherosclerosis, with a continuously increasing incidence in recent years, is the leading cause of coronary heart disease and stroke by plaque rupture and intraplaque hemorrhage. Vascular calcification, a process very much alike with osteogenesis, is considered to be a marker of advanced atherosclerosis. New evidence, suggesting the role of dietary intake influence on the diversity of the gut microbiome in the development of vascular calcifications, is highly debated. Gut microbiota can metabolize choline, phosphatidylcholine, and L-carnitine and produce vasculotoxic metabolites, such as trimethylamine-N-oxide (TMAO), a proatherogenic metabolite. This review article aims to discuss the latest research about how probiotics and the correction of diet is impacting the gut microbiota and its metabolites in the atherosclerotic process and vascular calcification. Further studies could create the premises for interventions in the microbiome as future primary tools in the prevention of atherosclerotic plaque and vascular calcifications.
Collapse
|
32
|
Li CY, Dai YX, Chang YT, Bai YM, Tsai SJ, Chen TJ, Chen MH. Proton Pump Inhibitors Are Associated with Increased Risk of Psoriasis: A Nationwide Nested Case-Control Study. Dermatology 2021; 237:884-890. [PMID: 34474411 DOI: 10.1159/000517515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/31/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are among the most widely used drugs. Little is known about the association between PPI use and risk of psoriasis. OBJECTIVE To investigate the association between PPI use and subsequent psoriasis risk. METHODS We included participants from the Taiwan National Health Insurance Research Database. Patients with PPI use and an incidence of psoriasis (n = 5,756) were assigned to the case cohort and 1:1 matched to controls. PPI use was defined as >30 cumulative defined daily doses (cDDDs); PPI nonuse was defined as ≤30 cDDDs. Logistic regression was used for the analyses. RESULTS There was a significant association between PPI use and psoriasis risk. The confounder-adjusted odd ratios (95% confidence interval [CI]) were 1.52 (1.31-1.76) and 1.54 (1.22-1.93) for patients with 120-365 cDDDs and >365 cDDDs, respectively, compared with PPI nonusers. Stratified analyses based on PPI type showed that exposure to lansoprazole (OR, 1.25; 95% CI, 1.11-1.41) was associated with subsequent psoriasis risk. CONCLUSIONS PPI use might be associated with an increased risk of developing psoriasis or as an epiphenomenon. Further prospective studies are needed to elucidate the association and underlying mechanisms.
Collapse
Affiliation(s)
- Cheng-Yuan Li
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Xiu Dai
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yun-Ting Chang
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Mei Bai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Jen Tsai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzeng-Ji Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
33
|
Wu H, Chiou J. Potential Benefits of Probiotics and Prebiotics for Coronary Heart Disease and Stroke. Nutrients 2021; 13:2878. [PMID: 34445037 PMCID: PMC8401746 DOI: 10.3390/nu13082878] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Among cardiovascular diseases (CVDs), a major cause of morbidity and mortality worldwide, coronary heart disease and stroke are the most well-known and extensively studied. The onset and progression of CVD is associated with multiple risk factors, among which, gut microbiota has received much attention in the past two decades. Gut microbiota, the microbial community colonizing in the gut, plays a prominent role in human health. In particular, gut dysbiosis is directly related to many acute or chronic dysfunctions of the cardiovascular system (CVS) in the host. Earlier studies have demonstrated that the pathogenesis of CVD is strongly linked to intestinal microbiota imbalance and inflammatory responses. Probiotics and prebiotics conferring various health benefits on the host are emerging as promising therapeutic interventions for many diseases. These two types of food supplements have the potential to alleviate the risks of CVD through improving the levels of several cardiovascular markers, such as total and low-density lipoprotein (LDL) cholesterol, high sensitivity C-reactive protein (hs-CRP), and certain cytokines involved in the inflammatory response. In this review, we focus mainly on the preventive effects of probiotics and prebiotics on CVD via rebalancing the structural and functional changes in gut microbiota and maintaining immune homeostasis.
Collapse
Affiliation(s)
- Haicui Wu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China;
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Jiachi Chiou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China;
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
34
|
Liu J, Xiao L, Nie H, Pan Y, Liu Y, Zhang Z, Lin X, Zhang Y, Cai J, Yang M, Liu Y, Zhang L, Xu A, Zhu C. Microecological preparation combined with an modified low-carbon diet improves glucolipid metabolism and cardiovascular complication in obese patients. Diabetol Metab Syndr 2021; 13:77. [PMID: 34256811 PMCID: PMC8276426 DOI: 10.1186/s13098-021-00697-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 07/02/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To investigate the impact of microecological preparation combined with modified low-carbon diet on the glucolipid metabolism and cardiovascular complication in obese patients. METHODS From August 2017 to July 2020, 66 obese patients were recruited, and administrated with an modified low-carbon diet with (group A) or without (Group B) microecology preparation and a balanced diet in control group (group C) for 6 months. Meanwhile, 20 volunteers administrated with a balanced diet were recruited as the healthy control group (group D). RESULTS After 6-month intervention, obese subjects in group A and B showed significant improvement of body and liver fat mass, reduction of serum lipid levels, intestinal barrier function markers, insulin resistance index (IRI), high blood pressure (HBP) and carotid intima thickness, as compared with subjects in group C. More importantly, subjects in group A had better improvement of vascular endothelial elasticity and intimal thickness than subjects in group B. However, these intervention had no effect on carotid atherosclerotic plaque. CONCLUSION Administration of microecological preparation combined with modified low-carbon diet had better improvement of intestinal barrier function, glucose and lipid metabolism, and cardiovascular complications than low-carbon diet in obese patients, but the effect of a simple low-carb diet on carotid atherosclerotic plaque need to be further addressed.
Collapse
Affiliation(s)
- Jianguo Liu
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Liehui Xiao
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Hezhongrong Nie
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Yong Pan
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University, Guangdong, Shenzhen, 518000, China
| | - Yan Liu
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Zhentian Zhang
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Xiuping Lin
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Yuan Zhang
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Jinchuang Cai
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Muxiu Yang
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Yajing Liu
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China
| | - Leijun Zhang
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China.
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Shenzhen, Hong Kong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Shenzhen, Hong Kong, China.
| | - Cuifeng Zhu
- Shenzhen Hospital of Southern Medical University, University of Hong Kong, Shenzhen, China.
- Department of Medicine, University of Hong Kong, Shenzhen, Hong Kong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Shenzhen, Hong Kong, China.
| |
Collapse
|
35
|
Chessa M, Panebianco M, Corbu S, Lussu M, Dessì A, Pintus R, Cesare Marincola F, Fanos V. Urinary Metabolomics Study of Patients with Bicuspid Aortic Valve Disease. Molecules 2021; 26:molecules26144220. [PMID: 34299495 PMCID: PMC8304733 DOI: 10.3390/molecules26144220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital heart defect responsible for valvular and aortic complications in affected patients. Causes and mechanisms of this pathology are still elusive and thus the lack of early detection biomarkers leads to challenges in its diagnosis and prevention of associated cardiovascular anomalies. The aim of this study was to explore the potential use of urine Nuclear Magnetic Resonance (NMR) metabolomics to evaluate a molecular fingerprint of BAV. Both multivariate and univariate statistical analyses were performed to compare the urinary metabolome of 20 patients with BAV with that of 24 matched controls. Orthogonal partial least squared discriminant analysis (OPLS-DA) showed statistically significant discrimination between cases and controls, suggesting seven metabolites (3-hydroxybutyrate, alanine, betaine, creatine, glycine, hippurate, and taurine) as potential biomarkers. Among these, glycine, hippurate and taurine individually displayed medium sensitivity and specificity by receiver operating characteristic (ROC) analysis. Pathway analysis indicated two metabolic pathways likely perturbed in BAV subjects. Possible contributions of gut microbiota activity and energy imbalance are also discussed. These results constitute encouraging preliminary findings in favor of the use of urine-based metabolomics for early diagnosis of BAV.
Collapse
Affiliation(s)
- Massimo Chessa
- Pediatric and Adult Congenital IRCCS, Policlinico San Donato, I-20097 San Donato Milanese, MI, Italy; (M.C.); (M.P.)
| | - Mario Panebianco
- Pediatric and Adult Congenital IRCCS, Policlinico San Donato, I-20097 San Donato Milanese, MI, Italy; (M.C.); (M.P.)
| | - Sara Corbu
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, University of Cagliari, S.P. n° 8, Km 0.700, I-09042 Monserrato, CA, Italy; (S.C.); (M.L.); (A.D.); (R.P.); (V.F.)
| | - Milena Lussu
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, University of Cagliari, S.P. n° 8, Km 0.700, I-09042 Monserrato, CA, Italy; (S.C.); (M.L.); (A.D.); (R.P.); (V.F.)
| | - Angelica Dessì
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, University of Cagliari, S.P. n° 8, Km 0.700, I-09042 Monserrato, CA, Italy; (S.C.); (M.L.); (A.D.); (R.P.); (V.F.)
| | - Roberta Pintus
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, University of Cagliari, S.P. n° 8, Km 0.700, I-09042 Monserrato, CA, Italy; (S.C.); (M.L.); (A.D.); (R.P.); (V.F.)
| | - Flaminia Cesare Marincola
- Department of Chemical and Geological Sciences, University of Cagliari, I-09042 Monserrato, CA, Italy
- Correspondence: ; Tel.: +39-070-675-4389
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria, University of Cagliari, S.P. n° 8, Km 0.700, I-09042 Monserrato, CA, Italy; (S.C.); (M.L.); (A.D.); (R.P.); (V.F.)
| |
Collapse
|
36
|
Liu Z, Liu H, Deng Q, Sun C, He W, Zheng W, Tang R, Li W, Xie Q. Association Between Dietary Inflammatory Index and Heart Failure: Results From NHANES (1999-2018). Front Cardiovasc Med 2021; 8:702489. [PMID: 34307508 PMCID: PMC8292138 DOI: 10.3389/fcvm.2021.702489] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: To explore the relationship between dietary inflammatory index (DII) and heart failure (HF) in participants with cardiovascular and cerebrovascular diseases. Methods: NHANES (1998–2018) data were collected and used to assess the association of HF with DII. Twenty-four-hour dietary consumptions were used to calculate the scores of DII. Demographic characteristics and physical and laboratory examinations were collected for the comparison between HF and non-HF groups. Logistic regression analysis and random forest analysis were performed to calculate the odds rate and determine the potential beneficial dietary components in HF. Results: A total of 19,067 cardiac-cerebral vascular disease participants were categorized as HF (n = 1,382; 7.25%) and non-HF (n = 17,685; 92.75%) groups. Heart failure participants had higher levels of DII score compared with those in the non-HF group (0.239 ± 1.702 vs. −0.145 ± 1.704, p < 0.001). Compared with individuals with T1 (DII: −3.884 to −0.570) of DII, those in T3 (DII: 1.019 to 4.598) had a higher level of total cholesterol (4.49 ± 1.16 vs. 4.75 ± 1.28 mmol/L, p < 0.01), globulin (29.92 ± 5.37 vs. 31.29 ± 5.84 g/L, p < 0.001), and pulse rate (69.90 ± 12.22 vs. 72.22 ± 12.77, p < 0.001) and lower levels of albumin (40.76 ± 3.52 vs. 39.86 ± 3.83 g/L, p < 0.001), hemoglobin (13.76 ± 1.65 vs. 13.46 ± 1.77 g/dl, p < 0.05), and hematocrit (40.83 ± 4.69 vs. 40.17 ± 5.01%, p < 0.05). The odds rates of HF for DII from the logistic regression were 1.140, 1.158, and 1.110 in models 1, 2, and 3, respectively. In addition, from the results of random forest analysis, dietary magnesium, fiber, and beta carotene may be essential in HF. Conclusion: Dietary inflammatory index was positively associated with HF in US adults, and dietary intervention might be a promising method in the therapy of HF.
Collapse
Affiliation(s)
- Zuheng Liu
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Haiyue Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Xiamen Key Laboratory of Genetic Testing, Xiamen, China
| | - Qinsheng Deng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Changqing Sun
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wangwei He
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wuyang Zheng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Rong Tang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Weihua Li
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qiang Xie
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
37
|
ACE2 as therapeutic agent. Clin Sci (Lond) 2021; 134:2581-2595. [PMID: 33063820 DOI: 10.1042/cs20200570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
The angiotensin-converting enzyme 2 (ACE2) has emerged as a critical regulator of the renin-angiotensin system (RAS), which plays important roles in cardiovascular homeostasis by regulating vascular tone, fluid and electrolyte balance. ACE2 functions as a carboxymonopeptidase hydrolyzing the cleavage of a single C-terminal residue from Angiotensin-II (Ang-II), the key peptide hormone of RAS, to form Angiotensin-(1-7) (Ang-(1-7)), which binds to the G-protein-coupled Mas receptor and activates signaling pathways that counteract the pathways activated by Ang-II. ACE2 is expressed in a variety of tissues and overwhelming evidence substantiates the beneficial effects of enhancing ACE2/Ang-(1-7)/Mas axis under many pathological conditions in these tissues in experimental models. This review will provide a succinct overview on current strategies to enhance ACE2 as therapeutic agent, and discuss limitations and future challenges. ACE2 also has other functions, such as acting as a co-factor for amino acid transport and being exploited by the severe acute respiratory syndrome coronaviruses (SARS-CoVs) as cellular entry receptor, the implications of these functions in development of ACE2-based therapeutics will also be discussed.
Collapse
|
38
|
Meikle SR, Sossi V, Roncali E, Cherry SR, Banati R, Mankoff D, Jones T, James M, Sutcliffe J, Ouyang J, Petibon Y, Ma C, El Fakhri G, Surti S, Karp JS, Badawi RD, Yamaya T, Akamatsu G, Schramm G, Rezaei A, Nuyts J, Fulton R, Kyme A, Lois C, Sari H, Price J, Boellaard R, Jeraj R, Bailey DL, Eslick E, Willowson KP, Dutta J. Quantitative PET in the 2020s: a roadmap. Phys Med Biol 2021; 66:06RM01. [PMID: 33339012 PMCID: PMC9358699 DOI: 10.1088/1361-6560/abd4f7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) plays an increasingly important role in research and clinical applications, catalysed by remarkable technical advances and a growing appreciation of the need for reliable, sensitive biomarkers of human function in health and disease. Over the last 30 years, a large amount of the physics and engineering effort in PET has been motivated by the dominant clinical application during that period, oncology. This has led to important developments such as PET/CT, whole-body PET, 3D PET, accelerated statistical image reconstruction, and time-of-flight PET. Despite impressive improvements in image quality as a result of these advances, the emphasis on static, semi-quantitative 'hot spot' imaging for oncologic applications has meant that the capability of PET to quantify biologically relevant parameters based on tracer kinetics has not been fully exploited. More recent advances, such as PET/MR and total-body PET, have opened up the ability to address a vast range of new research questions, from which a future expansion of applications and radiotracers appears highly likely. Many of these new applications and tracers will, at least initially, require quantitative analyses that more fully exploit the exquisite sensitivity of PET and the tracer principle on which it is based. It is also expected that they will require more sophisticated quantitative analysis methods than those that are currently available. At the same time, artificial intelligence is revolutionizing data analysis and impacting the relationship between the statistical quality of the acquired data and the information we can extract from the data. In this roadmap, leaders of the key sub-disciplines of the field identify the challenges and opportunities to be addressed over the next ten years that will enable PET to realise its full quantitative potential, initially in research laboratories and, ultimately, in clinical practice.
Collapse
Affiliation(s)
- Steven R Meikle
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Canada
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Richard Banati
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
- Australian Nuclear Science and Technology Organisation, Sydney, Australia
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, United States of America
| | - Terry Jones
- Department of Radiology, University of California, Davis, United States of America
| | - Michelle James
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), CA, United States of America
- Department of Neurology and Neurological Sciences, Stanford University, CA, United States of America
| | - Julie Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Internal Medicine, University of California, Davis, CA, United States of America
| | - Jinsong Ouyang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Chao Ma
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Suleman Surti
- Department of Radiology, University of Pennsylvania, United States of America
| | - Joel S Karp
- Department of Radiology, University of Pennsylvania, United States of America
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Taiga Yamaya
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Go Akamatsu
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Georg Schramm
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Ahmadreza Rezaei
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Johan Nuyts
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Roger Fulton
- Brain and Mind Centre, The University of Sydney, Australia
- Department of Medical Physics, Westmead Hospital, Sydney, Australia
| | - André Kyme
- Brain and Mind Centre, The University of Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Australia
| | - Cristina Lois
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Hasan Sari
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Julie Price
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, location VUMC, Netherlands
| | - Robert Jeraj
- Departments of Medical Physics, Human Oncology and Radiology, University of Wisconsin, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Slovenia
| | - Dale L Bailey
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Enid Eslick
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Kathy P Willowson
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Joyita Dutta
- Department of Electrical and Computer Engineering, University of Massachusetts Lowell, United States of America
| |
Collapse
|
39
|
Zhang R, Ma WQ, Fu MJ, Li J, Hu CH, Chen Y, Zhou MM, Gao ZJ, He YL. Overview of bile acid signaling in the cardiovascular system. World J Clin Cases 2021; 9:308-320. [PMID: 33521099 PMCID: PMC7812903 DOI: 10.12998/wjcc.v9.i2.308] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Bile acids (BAs) are classically known to play a vital role in the metabolism of lipids and in absorption. It is now well established that BAs act as signaling molecules, activating different receptors (such as farnesoid X receptor, vitamin D receptor, Takeda G-protein-coupled receptor 5, sphingosine-1-phosphate, muscarinic receptors, and big potassium channels) and participating in the regulation of energy homeostasis and lipid and glucose metabolism. In addition, increased BAs can impair cardiovascular function in liver cirrhosis. Approximately 50% of patients with cirrhosis develop cirrhotic cardiomyopathy. Exposure to high concentrations of hydrophobic BAs has been shown to be related to adverse effects with respect to vascular tension, endothelial function, arrhythmias, coronary atherosclerotic heart disease, and heart failure. The BAs in the serum BA pool have relevant through their hydrophobicity, and the lipophilic BAs are more harmful to the heart. Interestingly, ursodeoxycholic acid is a hydrophilic BA, and it is used as a therapeutic drug to reverse and protect the harmful cardiac effects caused by hydrophobic elevated BAs. In order to elucidate the mechanism of BAs and cardiovascular function, abundant experiments have been conducted in vitro and in vivo. The aim of this review was to explore the mechanism of BAs in the cardiovascular system.
Collapse
Affiliation(s)
- Rou Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Wen-Qi Ma
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Meng-Jun Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Chun-Hua Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Yi Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Mi-Mi Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Zhi-Jie Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Ying-Li He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
40
|
Chen C, Niu M, Pan J, Du N, Liu S, Li H, He Q, Mao J, Duan Y, Du Y. Bacteroides, butyric acid and t10,c12-CLA changes in colorectal adenomatous polyp patients. Gut Pathog 2021; 13:1. [PMID: 33436066 PMCID: PMC7805033 DOI: 10.1186/s13099-020-00395-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Colorectal adenomatous polyps (CAPs) are considered precancerous lesions of colorectal cancer (CRC). The gut microbiota participates in the process of digestion and, in the process, produces metabolites, mainly short-chain fatty acids (SCFAs), secondary bile acids and conjugated linoleic acid (CLA). This study aimed to investigate the gut microbiota constituents and metabolites in the faeces of CAP patients to identify microbiota or metabolites that can be used as sensitive biological predictors and to provide a theoretical basis for the clinical treatment of CAPs. METHODS 16S rRNA sequence analysis was used to detect microbial changes in the faeces of CAP patients. qPCR analysis was used to evaluate the ability of the microbiota to produce metabolites, and the contents of metabolites in faeces were detected by ion chromatography and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). RESULTS Based on the detection of the gut microbiota, patients with CAPs had increased abundances of Bacteroides and Citrobacter, and the abundances of Weissella and Lactobacillus were decreased. We also explored gene expression, and the abundance of butyrate-producing bacterial genes was significantly increased in the faeces of CAP patients, but those of secondary bile acid-producing and CLA-producing bacterial genes showed no differences in faecal samples. The acetic acid and butyric acid contents were increased in the faeces of the CAP group, and the healthy control group had higher t10,c12-CLA contents. CONCLUSION The gut microbiota analysis results, assessed in faeces, showed that Bacteroides and Citrobacter were positively correlated with CAPs, which indicated that changes in specific genera might be detrimental to intestinal health. In addition, t10,c12-CLA played an important role in protecting the intestine.
Collapse
Affiliation(s)
- Ciyan Chen
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China.,Qujing Healthcare Security Administration, Qujing, 655000, China
| | - Min Niu
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Junxi Pan
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Na Du
- Department of Clinical Laboratory, the No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Shumin Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Huanqin Li
- Department of Clinical Laboratory, the No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Qiuyue He
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Jian Mao
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Yong Duan
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China.,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
| | - Yan Du
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, 650032, China. .,Yunnan Institute of Laboratory Diagnosis, Kunming, 650032, China. .,Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China.
| |
Collapse
|
41
|
Zawada A, Rychter AM, Ratajczak AE, Lisiecka-Masian A, Dobrowolska A, Krela-Kaźmierczak I. Does Gut-Microbiome Interaction Protect against Obesity and Obesity-Associated Metabolic Disorders? Microorganisms 2020; 9:18. [PMID: 33374597 PMCID: PMC7822472 DOI: 10.3390/microorganisms9010018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
More research has recently focused on the role of the gut microbiota in the development or course of numerous diseases, including non-communicable diseases. As obesity remains prevalent, the question arises as to what microbial changes are associated with increased obesity prevalence and what kind of prevention and treatment approaches it could provide. Moreover, the influence of the gut-brain axis on obesity is also crucial, since it can affect metabolism and food intake. The quantitative and qualitative changes in the microbiota composition are called dysbiosis; however, in view of the current knowledge, it is difficult to conclude which microbial imbalances are adverse or beneficial. Increased numbers of pathological microorganisms were observed among patients with obesity and comorbidities associated with it, such as diabetes, cardiovascular disease, and insulin resistance. Our review provides current knowledge regarding changes in the intestinal microbiota associated with obesity and obesity-associated comorbidities. Nevertheless, given that dietary patterns and nutrients are two of the factors affecting the intestinal microbiota, we also discuss the role of different dietary approaches, vitamins, and minerals in the shaping of the intestinal microbiota.
Collapse
Affiliation(s)
| | - Anna Maria Rychter
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 49 Przybyszewskiego Street, 60-355 Poznan, Poland; (A.Z.); (A.E.R.); (A.L.-M.); (A.D.); (I.K.-K.)
| | | | | | | | | |
Collapse
|
42
|
Tabucanon T, Wilcox J, Tang WHW. Does Weight Loss Improve Clinical Outcomes in Overweight and Obese Patients with Heart Failure? Curr Diab Rep 2020; 20:75. [PMID: 33231788 DOI: 10.1007/s11892-020-01367-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Obesity increases the risk of new onset heart failure (HF), and particularly HF with preserved ejection fraction (HFpEF). Despite the observations of favorable clinical outcomes in HF patients with obesity in general, sometimes referred to as the "obesity paradox," it is important to recognize that severe obesity is associated with worse clinical outcomes. This review summarizes the effects of obesity treatment on cardiovascular health and HF clinical outcomes. RECENT FINDINGS Treatment for obesity utilizes a variety of modalities to achieve purposeful weight loss including lifestyle intervention, medications, and bariatric surgery. There are a cluster of benefits of obesity treatment in terms of clinical outcomes in HF. The mechanisms of these benefits include both weight loss-dependent and weight loss-independent mechanisms. Obesity treatment is safe and associated with favorable clinical outcomes across the spectrum of the HF population. The potential benefits are facilitated through multiple mechanisms.
Collapse
Affiliation(s)
- Thida Tabucanon
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Kaufman Center for Heart Failure Treatment and Recovery, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA
- Thammasat Heart Center, Thammasat University Hospital, Khlong Luang, Pathum Thani, Thailand
| | - Jennifer Wilcox
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Kaufman Center for Heart Failure Treatment and Recovery, Cleveland Clinic, 9500 Euclid Avenue, Desk J3-4, Cleveland, OH, 44195, USA.
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
43
|
How Periodontal Disease and Presence of Nitric Oxide Reducing Oral Bacteria Can Affect Blood Pressure. Int J Mol Sci 2020; 21:ijms21207538. [PMID: 33066082 PMCID: PMC7589924 DOI: 10.3390/ijms21207538] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO), a small gaseous and multifunctional signaling molecule, is involved in the maintenance of metabolic and cardiovascular homeostasis. It is endogenously produced in the vascular endothelium by specific enzymes known as NO synthases (NOSs). Subsequently, NO is readily oxidized to nitrite and nitrate. Nitrite is also derived from exogenous inorganic nitrate (NO3) contained in meat, vegetables, and drinking water, resulting in greater plasma NO2 concentration and major reduction in systemic blood pressure (BP). The recycling process of nitrate and nitrite to NO (nitrate-nitrite-NO pathway), known as the enterosalivary cycle of nitrate, is dependent upon oral commensal nitrate-reducing bacteria of the dorsal tongue. Veillonella, Actinomyces, Haemophilus, and Neisseria are the most copious among the nitrate-reducing bacteria. The use of chlorhexidine mouthwashes and tongue cleaning can mitigate the bacterial nitrate-related BP lowering effects. Imbalances in the oral reducing microbiota have been associated with a decrease of NO, promoting endothelial dysfunction, and increased cardiovascular risk. Although there is a relationship between periodontitis and hypertension (HT), the correlation between nitrate-reducing bacteria and HT has been poorly studied. Restoring the oral flora and NO activity by probiotics may be considered a potential therapeutic strategy to treat HT.
Collapse
|
44
|
Matsumoto T, Kojima M, Takayanagi K, Taguchi K, Kobayashi T. Trimethylamine-N-oxide Specifically Impairs Endothelium-Derived Hyperpolarizing Factor-Type Relaxation in Rat Femoral Artery. Biol Pharm Bull 2020; 43:569-573. [PMID: 32115516 DOI: 10.1248/bpb.b19-00957] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although substantial evidence suggests that an increase in the level of trimethylamine-N-oxide (TMAO) is associated with the risk of cardiovascular diseases, including atherosclerosis, chronic kidney diseases, and hypertension, the direct effect of TMAO on vascular endothelial function remains unclear. Therefore, we investigated the acute effects of TMAO on endothelium-dependent relaxation induced by acetylcholine (ACh) in the superior mesenteric arteries and femoral arteries of rat. In endothelium-intact preparations, it was observed that TMAO (300 µmol/L for 60 min) did not affect ACh-induced relaxation in either of the two arteries. In endothelium-derived hyperpolarizing factor (EDHF)-mediated relaxation under nitric oxide synthase (NOS) and cyclooxygenase (COX) inhibitions by Nω-nitro-L-arginine (L-NNA) and indomethacin, respectively, TMAO specifically impairs the relaxation in femoral arteries but not in the superior mesenteric arteries. Under the inhibitory actions of NOS and as well as blockade of intermediate-conductance calcium-activated potassium channel (IKCa) (by TRAM-34) and small-conductance calcium-activated potassium channel (SKCa) (by apamin), which are putative sources of EDHF, ACh-induced relaxation was low, and there were no differences between the control and TMAO-treated groups with respect to both arteries. In femoral arteries, TMAO slightly reduces ACh-induced relaxation in the presence of indomethacin (preserved NO and EDHF signals) but does not affect ACh-induced NO-mediated relaxation under the combined presence of indomethacin, TRAM-34, and apamin. These results suggest that acute treatment with TMAO specifically impairs EDHF-mediated relaxation in the femoral arteries but not in the superior mesenteric arteries. These novel observations show that TMAO is a causative factor in the development of peripheral arterial disease.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Mihoka Kojima
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Keisuke Takayanagi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| |
Collapse
|
45
|
Gutiérrez-Calabrés E, Ortega-Hernández A, Modrego J, Gómez-Gordo R, Caro-Vadillo A, Rodríguez-Bobada C, González P, Gómez-Garre D. Gut Microbiota Profile Identifies Transition From Compensated Cardiac Hypertrophy to Heart Failure in Hypertensive Rats. Hypertension 2020; 76:1545-1554. [PMID: 32921194 DOI: 10.1161/hypertensionaha.120.15123] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Microcirculatory alterations displayed by patients with heart failure (HF) induce structural and functional intestinal changes that may affect normal gut microbial community. At the same time, gut microbiota can influence pathological mechanisms implicated in HF progression. However, it is unknown whether gut microbiota dysbiosis can precede the development of cardiac alterations in HF or it is only a mere consequence. Our aim was to investigate the potential relationship between gut microbiota composition and HF development by comparing spontaneously hypertensive heart failure and spontaneously hypertensive rat models. Gut microbiota from spontaneously hypertensive heart failure, spontaneously hypertensive rat, and normotensive Wistar Kyoto rats at 9 and 19 months of age was analyzed by sequencing the 16S ribosomal RNA gene, and KEGG metabolic pathways associated to 16S profiles were predicted. Beta diversity, Firmicutes/Bacteroidetes ratio, taxonomic abundances, and potential metabolic functions of gut microbiota were significantly different in spontaneously hypertensive heart failure with respect to spontaneously hypertensive rat before (9 months) and after (19 months) cardiac differences were presented. Nine-month-old spontaneously hypertensive heart failure showed a significant increase in the genera Paraprevotella, Oscillospira, Prevotella 9, Faecalitalea, Faecalibacterium, Ruminiclostridium 6, Phascolarctobacterium, Butyrivibrio, Parasutterella, and Parabacteroides compared with both Wistar Kyoto and spontaneously hypertensive rat, while Ruminiclostridium 9, Oscillibacter, Ruminiclostridium, Mucispirillum, Intestinimonas, and Akkermansia were diminished. Of them, Akkermansia, Prevotella 9, Paraprevotella, and Phascolarctobaterium were associated to changes in cardiac structure and function. Our results demonstrate an association between specific changes in gut microbiota and the development of HF in a hypertensive model of HF and further support the intervention to restore gut microbiota as an innovative therapeutic strategy for preventing HF.
Collapse
Affiliation(s)
- Elena Gutiérrez-Calabrés
- From the Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain (E.G.-C., A.O.-H., J.M., R.G.-G., D.G.-G.)
| | - Adriana Ortega-Hernández
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain (J.M., D.G.-G.)
| | - Javier Modrego
- From the Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain (E.G.-C., A.O.-H., J.M., R.G.-G., D.G.-G.)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain (J.M., D.G.-G.)
| | - Rubén Gómez-Gordo
- From the Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain (E.G.-C., A.O.-H., J.M., R.G.-G., D.G.-G.)
| | - Alicia Caro-Vadillo
- Department of Medicine and Animal Surgery, Veterinary School, Universidad Complutense, Madrid, Spain (A.C.-V.)
| | - Cruz Rodríguez-Bobada
- Unit of Medicine and Experimental Surgery, Hospital Clínico San Carlos-IdISSC, Madrid, Spain (C.R.-B., P.G.)
| | - Pablo González
- Unit of Medicine and Experimental Surgery, Hospital Clínico San Carlos-IdISSC, Madrid, Spain (C.R.-B., P.G.)
| | - Dulcenombre Gómez-Garre
- From the Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain (E.G.-C., A.O.-H., J.M., R.G.-G., D.G.-G.)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain (J.M., D.G.-G.)
| |
Collapse
|
46
|
Sikora M, Stec A, Chrabaszcz M, Knot A, Waskiel-Burnat A, Rakowska A, Olszewska M, Rudnicka L. Gut Microbiome in Psoriasis: An Updated Review. Pathogens 2020; 9:pathogens9060463. [PMID: 32545459 PMCID: PMC7350295 DOI: 10.3390/pathogens9060463] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: A growing body of evidence highlights that intestinal dysbiosis is associated with the development of psoriasis. The gut–skin axis is the novel concept of the interaction between skin diseases and microbiome through inflammatory mediators, metabolites and the intestinal barrier. The objective of this study was to synthesize current data on the gut microbial composition in psoriasis. (2) Methods: We conducted a systematic review of studies investigating intestinal microbiome in psoriasis, using the PRISMA checklist. We searched MEDLINE, EMBASE, and Web of Science databases for relevant published articles (2000–2020). (3) Results: All of the 10 retrieved studies reported alterations in the gut microbiome in patients with psoriasis. Eight studies assessed alpha- and beta-diversity. Four of them reported a lack of change in alpha-diversity, but all confirmed significant changes in beta-diversity. At the phylum-level, at least two or more studies reported a lower relative abundance of Bacteroidetes, and higher Firmicutes in psoriasis patients versus healthy controls. (4) Conclusions: There is a significant association between alterations in gut microbial composition and psoriasis; however, there is high heterogeneity between studies. More unified methodological standards in large-scale studies are needed to understand microbiota’s contribution to psoriasis pathogenesis and its modulation as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
- Correspondence:
| | - Albert Stec
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Magdalena Chrabaszcz
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Aleksandra Knot
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Anna Waskiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Adriana Rakowska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Malgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| |
Collapse
|
47
|
Moludi J, Maleki V, Jafari‐Vayghyan H, Vaghef‐Mehrabany E, Alizadeh M. Metabolic endotoxemia and cardiovascular disease: A systematic review about potential roles of prebiotics and probiotics. Clin Exp Pharmacol Physiol 2020; 47:927-939. [DOI: 10.1111/1440-1681.13250] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/28/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Jalal Moludi
- School of Nutrition Sciences and Food Technology Kermanshah University of Medical Sciences Kermanshah Iran
- Clinical Research Development Center Imam Reza Hospital Kermanshah University of Medical Sciences Kermanshah Iran
| | - Vahid Maleki
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | | | - Elnaz Vaghef‐Mehrabany
- Nutrition Research Center Faculty of Nutrition Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Alizadeh
- Nutrition Research Center Faculty of Nutrition Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
48
|
Formiga F, Ferreira Teles CI, Chivite D. Impact of intestinal microbiota in patients with heart failure: A systematic review. Med Clin (Barc) 2019; 153:402-409. [PMID: 31416611 DOI: 10.1016/j.medcli.2019.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 10/26/2022]
Abstract
Heart failure (HF) is a chronic disease with significant morbidity and mortality. Substantial haemodynamic changes such as hypoperfusion and intestinal congestion can alter the composition of the intestinal microbiota in patients with HF. The aim of this systematic review is to evaluate the influence of bowel function in patients with HF and the possible role of the intestinal microbiota in the development and evolution of the latter. Eleven studies were included in the review. These studies seem to confirm that HF patients present with substantial abnormalities in the composition of their intestinal microbiota. Trimethylamine N-oxide is identified as a key mediator between the alterations in the intestinal microbiota and HF and correlates with worse prognosis in HF patients. In conclusion, patients with HF present with frequent abnormalities in the characteristics of their intestinal microbiota, which may play a role in the prognosis of the disease.
Collapse
Affiliation(s)
- Francesc Formiga
- Servicio de Medicina Interna, Hospital Universitario de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, España.
| | - Cristiana Isabel Ferreira Teles
- Servicio de Medicina Interna, Hospital Universitario de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, España
| | - David Chivite
- Servicio de Medicina Interna, Hospital Universitario de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, España
| |
Collapse
|
49
|
Thenappan T, Khoruts A, Chen Y, Weir EK. Can intestinal microbiota and circulating microbial products contribute to pulmonary arterial hypertension? Am J Physiol Heart Circ Physiol 2019; 317:H1093-H1101. [PMID: 31490732 DOI: 10.1152/ajpheart.00416.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease with a median survival of only 5-7 yr. PAH is characterized by remodeling of the pulmonary vasculature causing reduced pulmonary arterial compliance (PAC) and increased pulmonary vascular resistance (PVR), ultimately resulting in right ventricular failure and death. Better therapies for PAH will require a paradigm shift in our understanding of the early pathophysiology. PAC decreases before there is an increase in the PVR. Unfortunately, present treatment has little effect on PAC. The loss of compliance correlates with extracellular matrix remodeling and fibrosis in the pulmonary vessels, which have been linked to chronic perivascular inflammation and immune dysregulation. However, what initiates the perivascular inflammation and immune dysregulation in PAH is unclear. Alteration of the gut microbiota composition and function underlies the level of immunopathogenic involvement in several diseases, including atherosclerosis, obesity, diabetes mellitus, and depression, among others. In this review, we discuss evidence that raises the possibility of an etiologic role for changes in the gut and circulating microbiome in the initiation of perivascular inflammation in the early pathogenesis of PAH.
Collapse
Affiliation(s)
- Thenappan Thenappan
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota.,BioTechnology Institute, University of Minnesota, Minneapolis, Minnesota
| | - Yingjie Chen
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - E Kenneth Weir
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
50
|
Ahmad AF, Dwivedi G, O'Gara F, Caparros-Martin J, Ward NC. The gut microbiome and cardiovascular disease: current knowledge and clinical potential. Am J Physiol Heart Circ Physiol 2019; 317:H923-H938. [PMID: 31469291 DOI: 10.1152/ajpheart.00376.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The human body is populated by a diverse community of microbes, dominated by bacteria, but also including viruses and fungi. The largest and most complex of these communities is located in the gastrointestinal system and, with its associated genome, is known as the gut microbiome. Gut microbiome perturbations and related dysbiosis have been implicated in the progression and pathogenesis of CVD, including atherosclerosis, hypertension, and heart failure. Although there have been advances in the characterization and analysis of the gut microbiota and associated bacterial metabolites, the exact mechanisms through which they exert their action are not well understood. This review will focus on the role of the gut microbiome and associated functional components in the development and progression of atherosclerosis. Potential treatments to alter the gut microbiome to prevent or treat atherosclerosis and CVD are also discussed.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia.,Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Fergal O'Gara
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,BIOMERIT Research Centre, School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.,Telethon Kids Institute, Children's Hospital, Perth, Western Australia, Australia
| | - Jose Caparros-Martin
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Natalie C Ward
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,School of Public Health, Curtin University, Perth Western Australia, Australia
| |
Collapse
|