1
|
Giakomidi D, Ishola A, Nus M. Targeting gut microbiota to regulate the adaptive immune response in atherosclerosis. Front Cardiovasc Med 2025; 12:1502124. [PMID: 39957996 PMCID: PMC11825770 DOI: 10.3389/fcvm.2025.1502124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Atherosclerosis, the leading cause of death worldwide, is a chronic inflammatory disease leading to the accumulation of lipid-rich plaques in the intima of large and medium-sized arteries. Accumulating evidence indicates the important regulatory role of the adaptive immune system in atherosclerosis during all stages of the disease. The gut microbiome has also become a key regulator of atherosclerosis and immunomodulation. Whilst existing research extensively explores the impact of the microbiome on the innate immune system, only a handful of studies have explored the regulatory capacity of the microbiome on the adaptive immune system to modulate atherogenesis. Building on these concepts and the pitfalls on the gut microbiota and adaptive immune response interaction, this review explores potential strategies to therapeutically target the microbiome, including the use of prebiotics and vaccinations, which could influence the adaptive immune response and consequently plaque composition and development.
Collapse
Affiliation(s)
- Despina Giakomidi
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| | - Ayoola Ishola
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Cardiovascular Division, Department of Medicine, Heart and Lung Research Institute (HLRI), University of Cambridge, Cambridge, United Kingdom
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
2
|
Iatcu OC, Hamamah S, Covasa M. Harnessing Prebiotics to Improve Type 2 Diabetes Outcomes. Nutrients 2024; 16:3447. [PMID: 39458444 PMCID: PMC11510484 DOI: 10.3390/nu16203447] [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: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiota, a complex ecosystem of microorganisms in the human gastrointestinal tract (GI), plays a crucial role in maintaining metabolic health and influencing disease susceptibility. Dysbiosis, or an imbalance in gut microbiota, has been linked to the development of type 2 diabetes mellitus (T2DM) through mechanisms such as reduced glucose tolerance and increased insulin resistance. A balanced gut microbiota, or eubiosis, is associated with improved glucose metabolism and insulin sensitivity, potentially reducing the risk of diabetes-related complications. Various strategies, including the use of prebiotics like inulin, fructooligosaccharides, galactooligosaccharides, resistant starch, pectic oligosaccharides, polyphenols, β-glucan, and Dendrobium officinale have been shown to improve gut microbial composition and support glycemic control in T2DM patients. These prebiotics can directly impact blood sugar levels while promoting the growth of beneficial bacteria, thus enhancing glycemic control. Studies have shown that T2DM patients often exhibit a decrease in beneficial butyrate-producing bacteria, like Roseburia and Faecalibacterium, and an increase in harmful bacteria, such as Escherichia and Prevotella. This review aims to explore the effects of different prebiotics on T2DM, their impact on gut microbiota composition, and the potential for personalized dietary interventions to optimize diabetes management and improve overall health outcomes.
Collapse
Affiliation(s)
- Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
3
|
Flori L, Benedetti G, Martelli A, Calderone V. Microbiota alterations associated with vascular diseases: postbiotics as a next-generation magic bullet for gut-vascular axis. Pharmacol Res 2024; 207:107334. [PMID: 39103131 DOI: 10.1016/j.phrs.2024.107334] [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: 05/14/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
The intestinal microbiota represents a key element in maintaining the homeostasis and health conditions of the host. Vascular pathologies and other risk factors such as aging have been recently associated with dysbiosis. The qualitative and quantitative alteration of the intestinal microbiota hinders correct metabolic homeostasis, causing structural and functional changes of the intestinal wall itself. Impairment of the intestinal microbiota, combined with the reduction of the barrier function, worsen the pathological scenarios of peripheral tissues over time, including the vascular one. Several experimental evidence, collected in this review, describes in detail the changes of the intestinal microbiota in dysbiosis associated with vascular alterations, such as atherosclerosis, hypertension, and endothelial dysfunction, the resulting metabolic disorders and how these can impact on vascular health. In this context, the gut-vascular axis is considered, for the first time, as a merged unit involved in the development and progression of vascular pathologies and as a promising target. Current approaches for the management of dysbiosis such as probiotics, prebiotics and dietary modifications act mainly on the intestinal district. Postbiotics, described as preparation of inanimate microorganisms and/or their components that confers health benefits on the host, represent an innovative strategy for a dual management of intestinal dysbiosis and vascular pathologies. In this context, this review has the further purpose of defining the positive effects of the supplementation of bacterial strains metabolites (short‑chain fatty acids, exopolysaccharides, lipoteichoic acids, gallic acid, and protocatechuic acid) restoring intestinal homeostasis and acting directly on the vascular district through the gut-vascular axis.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Giada Benedetti
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| |
Collapse
|
4
|
Forss A, Ma W, Thuresson M, Sun J, Ebrahimi F, Bergman D, Olén O, Sundström J, Ludvigsson JF. Diverticular disease and risk of incident major adverse cardiovascular events: A nationwide matched cohort study. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2024:qcae074. [PMID: 39174483 DOI: 10.1093/ehjqcco/qcae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
BACKGROUND An increased risk of cardiovascular disease (CVD) has been reported in patients with diverticular disease (DD). However, there are knowledge gaps about specific risks of each major adverse cardiovascular event (MACE) component. METHODS This nationwide cohort study included Swedish adults with DD (1987-2017, N=52,468) without previous CVD. DD was defined through ICD codes in the National Patient Register and colorectal histopathology reports from the ESPRESSO study. DD cases were matched by age, sex, calendar year and county of residence to ≤5 population reference individuals (N=194,525). Multivariable-adjusted hazard ratios (aHRs) for MACE up until December 2021 were calculated using stratified Cox proportional hazard models. RESULTS Median age at DD diagnosis was 62 years and 61% were females. During a median follow-up of 8.6 years, 16,147 incident MACE occurred in individuals with DD, and 48,134 in reference individuals (incidence rates (IRs)=61.4 vs. 43.8/1,000 person-years) corresponding to an aHR of 1.24 (95%CI=1.22-1.27), equivalent to one extra case of MACE for every 6 DD patients followed for 10 years. The risk was increased for ischemic heart disease (IR=27.9 vs. 18.6; aHR=1.36, 95%CI=1.32-1.40), congestive heart failure (IR=23.2 vs. 15.8; aHR=1.26, 95%CI=1.22-1.31), and stroke (IR=18.0 vs. 13.7; aHR=1.15, 95%CI=1.11-1.19). DD was not associated with cardiovascular mortality (IR=18.9 vs. 15.3; aHR=1.01, 95%CI=0.98-1.05). Results remained robust in sibling-controlled analyses. CONCLUSIONS Patients with DD had a 24% increased risk of MACE compared with reference individuals, but no increased cardiovascular mortality. Future research should confirm these data and examine underlying mechanisms and shared risk factors between DD and CVD.
Collapse
Affiliation(s)
- Anders Forss
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Gastroenterology Unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marcus Thuresson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Statisticon AB, Sweden
| | - Jiangwei Sun
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fahim Ebrahimi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology and Hepatology, University Digestive Health Care Center Basel - Clarunis, Basel, Switzerland
| | - David Bergman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ola Olén
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Stockholm South General Hospital, Stockholm, Sweden
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
5
|
Völkerer A, Wernly S, Semmler G, Flamm M, Radzikowski K, Datz L, Götz N, Hofer H, Aigner E, Datz C, Wernly B. Association between Cardiovascular Risk Assessment by SCORE2 and Diverticulosis: A Cross-Sectional Analysis. J Pers Med 2024; 14:862. [PMID: 39202053 PMCID: PMC11355319 DOI: 10.3390/jpm14080862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND The aim of this retrospective observational study was to examine the relationship between SCORE2 and the occurrence of colonic diverticula in a screening population without cardiovascular or gastrointestinal symptoms. SCORE2, recognized and supported by the European Society of Cardiology for cardiovascular risk assessment, served as the primary metric for the analysis in this investigation. METHODS We studied 3935 asymptomatic individuals undergoing screening colonoscopy. SCORE2 was calculated for each participant and categorized into three groups based on the following projected 10-year cardiovascular disease risk: SCORE2 0-4.9%, SCORE2 5-9.9%, and SCORE2 ≥ 10%. Logistic regression was used to assess the relationship between SCORE2 and diverticulosis. RESULTS SCORE2 was associated with the presence of diverticulosis (OR 1.09, 95%CI 1.07-1.10; p < 0.001) in univariable logistic regression, translating into an RR of 1.07 per unit increase. The association persisted after multivariable adjusting for metabolic syndrome (aOR 1.08; 95%CI 1.06-1.10; p < 0.001). Patients with high cardiovascular risk had higher rates of diverticulosis compared to those with lower risk: high risk (OR 2.00, 95%CI 1.71-2.33; p < 0.001); very high risk (OR 2.53, 95%CI 2.10-3.05; p < 0.001). This association remained after adjusting for metabolic syndrome: high risk (aOR 1.86, 95%CI 1.59-2.18; p < 0.001); very high risk (aOR 2.27, 95%CI 1.88-2.75; p < 0.001). CONCLUSIONS A higher SCORE2 was found to be a suitable screening parameter for diverticular disease. This suggests a potential link between cardiovascular risk factors and colon diverticula development, warranting further research on whether optimizing cardiovascular risk factors could positively influence diverticular disease.
Collapse
Affiliation(s)
- Andreas Völkerer
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Sarah Wernly
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Maria Flamm
- Institute of General Practice, Family Medicine and Preventive Medicine, Center for Public Health and Healthcare Research Paracelsus Medical University, 5020 Salzburg, Austria
| | - Konrad Radzikowski
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Leonora Datz
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Nikolaus Götz
- Department of Internal Medicine, General Hospital St. Vinzenz Zams, 6511 Zams, Austria
| | - Hannah Hofer
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Elmar Aigner
- Clinic I for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Christian Datz
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Bernhard Wernly
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
- Institute of General Practice, Family Medicine and Preventive Medicine, Center for Public Health and Healthcare Research Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
6
|
Naghipour S, Cox AJ, Fisher JJ, Plan M, Stark T, West N, Peart JN, Headrick JP, Du Toit EF. Circulating TMAO, the gut microbiome and cardiometabolic disease risk: an exploration in key precursor disorders. Diabetol Metab Syndr 2024; 16:133. [PMID: 38886825 PMCID: PMC11181661 DOI: 10.1186/s13098-024-01368-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Elevations in the gut metabolite trimethylamine-N-oxide (TMAO) have been linked to cardiovascular and metabolic diseases. Whether elevated TMAO levels reflect early mechanistic involvement or a sequela of evolving disease awaits elucidation. The purpose of this study was to further explore these potential associations. METHODS We investigated relationships between circulating levels of TMAO and its pre-cursor substrates, dietary factors, gut microbiome profiles and disease risk in individuals with a Healthy BMI (18.5 < BMI < 25, n = 41) or key precursor states for cardiometabolic disease: Overweight (25 < BMI < 30 kg/m2, n = 33), Obese (BMI > 30, n = 27) and Metabolic Syndrome (MetS; ≥ 3 ATPIII report criteria, n = 39). RESULTS Unexpectedly, plasma [TMAO] did not vary substantially between groups (means of 3-4 µM; p > 0.05), although carnitine was elevated in participants with MetS. Gut microbial diversity and Firmicutes were also significantly reduced in the MetS group (p < 0.05). Exploratory analysis across diverse parameters reveals significant correlations between circulating [TMAO] and seafood intake (p = 0.007), gut microbial diversity (p = 0.017-0.048), and plasma [trimethylamine] (TMA; p = 0.001). No associations were evident with anthropometric parameters or cardiometabolic disease risk. Most variance in [TMAO] within and between groups remained unexplained. CONCLUSIONS Data indicate that circulating [TMAO] may be significantly linked to seafood intake, levels of TMA substrate and gut microbial diversity across healthy and early disease phenotypes. However, mean concentrations remain < 5 µM, with little evidence of links between TMAO and cardiometabolic disease risk. These observations suggest circulating TMAO may not participate mechanistically in cardiometabolic disease development, with later elevations likely a detrimental sequela of extant disease.
Collapse
Affiliation(s)
- Saba Naghipour
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, 4215, Australia
| | - Amanda J Cox
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, 4215, Australia
- Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, QLD, 4215, Australia
| | - Joshua J Fisher
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Manuel Plan
- Metabolomics Australia (Queensland Node), The University of Queensland, St. Lucia, QLD, 4072, Australia
- Metabolomics Facility, QIMR Berghofer Medical Research Institute, 300 Herston Rd, Herston, QLD, 4006, Australia
| | - Terra Stark
- Metabolomics Australia (Queensland Node), The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Nic West
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, 4215, Australia
- Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, QLD, 4215, Australia
| | - Jason N Peart
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, 4215, Australia
| | - John P Headrick
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, 4215, Australia
| | - Eugene F Du Toit
- School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, 4215, Australia.
| |
Collapse
|
7
|
Li X, Li Q, Wang L, Ding H, Wang Y, Liu Y, Gong T. The interaction between oral microbiota and gut microbiota in atherosclerosis. Front Cardiovasc Med 2024; 11:1406220. [PMID: 38932989 PMCID: PMC11199871 DOI: 10.3389/fcvm.2024.1406220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis (AS) is a complex disease caused by multiple pathological factors threatening human health-the pathogenesis is yet to be fully elucidated. In recent years, studies have exhibited that the onset of AS is closely involved with oral and gut microbiota, which may initiate or worsen atherosclerotic processes through several mechanisms. As for how the two microbiomes affect AS, existing mechanisms include invading plaque, producing active metabolites, releasing lipopolysaccharide (LPS), and inducing elevated levels of inflammatory mediators. Considering the possible profound connection between oral and gut microbiota, the effect of the interaction between the two microbiomes on the initiation and progression of AS has been investigated. Findings are oral microbiota can lead to gut dysbiosis, and exacerbate intestinal inflammation. Nevertheless, relevant research is not commendably refined and a concrete review is needed. Hence, in this review, we summarize the most recent mechanisms of the oral microbiota and gut microbiota on AS, illustrate an overview of the current clinical and epidemiological evidence to support the bidirectional connection between the two microbiomes and AS.
Collapse
Affiliation(s)
- Xinsi Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Municipal KeyLaboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Qian Li
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Municipal KeyLaboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Li Wang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Municipal KeyLaboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Department of Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Huifen Ding
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Municipal KeyLaboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Department of Prosthodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yizhong Wang
- Department of Research & Development, Zhejiang Charioteer Pharmaceutical Co., Ltd, Taizhou, China
| | - Yunfei Liu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Municipal KeyLaboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Department of Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Gong
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Municipal KeyLaboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Department of Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Borsinger T, Torchia M, Malskis B, Levy BA, Werth PM, Moschetti WE. Characterizing the Native Microbiome Using Next-Generation Sequencing of Bilateral 'Aseptic' Knees. J Arthroplasty 2024; 39:1317-1322. [PMID: 37952737 DOI: 10.1016/j.arth.2023.11.002] [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: 06/09/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Next generation sequencing (NGS) has proven ability to identify organisms beyond those identified through traditional culture-based techniques in cases of suspected prosthetic joint infection. However, there is concern that some microorganisms identified may represent the natural joint microbiome rather than pathogenic agents. This work sought to evaluate the presence of microorganisms identified with NGS in bilateral native, presumed "aseptic" knees with osteoarthritis. METHODS There were 40 patients undergoing primary unilateral (30) or bilateral (10) total knee arthroplasty enrolled prospectively. During surgery, samples of fluid and tissue were obtained from operative knees, and joint fluid was obtained from nonoperative knees. Samples were sent for NGS analysis and processed according to manufacturer protocols. Patient age, body mass index, comorbidities, prior history of injections, and grade of arthritis were evaluated for association with positive NGS results. RESULTS There were 3 of 80 samples (3.8%) that demonstrated positive NGS. There were two of these that had multiple microorganisms identified (1 knee with 4 microorganisms; 1 knee with 2 microorganisms). An additional 2 samples had positive NGS results below the manufacturer's threshold for reporting. The most common organism identified was Cutibacterium acnes, present in 2 of the 3 positive samples. No patient baseline characteristics were associated with positive NGS results. CONCLUSIONS Some native knee joints with osteoarthritis have positive microorganisms identified with NGS. The presence of microorganisms in the native knee has important implications for better understanding the native joint microbiome as well as utilization of NGS in cases of suspected prosthetic joint infection.
Collapse
Affiliation(s)
- Tracy Borsinger
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Michael Torchia
- Department of Orthopaedics, Essentia Health Duluth Clinic, Duluth, Minnesota
| | - Bethany Malskis
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Benjamin A Levy
- Department of Orthopaedics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Paul M Werth
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Wayne E Moschetti
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| |
Collapse
|
9
|
Wang J, Hu Z, Xu Q, Shi Y, Cao X, Ma Y, Wang M, Zhang C, Luo X, Lin F, Li X, Duan Y, Cai H. Gut microbiome-based noninvasive diagnostic model to predict acute coronary syndromes. Front Cell Infect Microbiol 2024; 13:1305375. [PMID: 38298920 PMCID: PMC10829574 DOI: 10.3389/fcimb.2023.1305375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Background Previous studies have shown that alterations in the gut microbiota are closely associated with Acute Coronary Syndrome (ACS) development. However, the value of gut microbiota for early diagnosis of ACS remains understudied. Methods We recruited 66 volunteers, including 29 patients with a first diagnosis of ACS and 37 healthy volunteers during the same period, collected their fecal samples, and sequenced the V4 region of the 16S rRNA gene. Functional prediction of the microbiota was performed using PICRUSt2. Subsequently, we constructed a nomogram and corresponding webpage based on microbial markers to assist in the diagnosis of ACS. The diagnostic performance and usefulness of the model were analyzed using boostrap internal validation, calibration curves, and decision curve analysis (DCA). Results Compared to that of healthy controls, the diversity and composition of microbial community of patients with ACS was markedly abnormal. Potentially pathogenic genera such as Streptococcus and Acinetobacter were significantly increased in the ACS group, whereas certain SCFA-producing genera such as Blautia and Agathobacter were depleted. In addition, in the correlation analysis with clinical indicators, the microbiota was observed to be associated with the level of inflammation and severity of coronary atherosclerosis. Finally, a diagnostic model for ACS based on gut microbiota and clinical variables was developed with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.963 (95% CI: 0.925-1) and an AUC value of 0.948 (95% CI: 0.549-0.641) for bootstrap internal validation. The calibration curves of the model show good consistency between the actual and predicted probabilities. The DCA showed that the model had a high net clinical benefit for clinical applications. Conclusion Our study is the first to characterize the composition and function of the gut microbiota in patients with ACS and healthy populations in Southwest China and demonstrates the potential effect of the microbiota as a non-invasive marker for the early diagnosis of ACS.
Collapse
Affiliation(s)
- Jincheng Wang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Hu
- Department of Geriatric Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qiuyue Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, China
| | - Yunke Shi
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingyu Cao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiming Ma
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mingqiang Wang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chaoyue Zhang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiang Luo
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fanru Lin
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xianbin Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, China
| | - Hongyan Cai
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
10
|
WEERARATHNA TP, LEKAMWASAM S, KODIKARA I, WASANA KGP, FONSEKA L. Control of cardiometabolic risk factors and their association with carotid intima media thickness among patients with type 2 diabetes mellitus-single center experience in a developing country. Turk J Med Sci 2024; 54:545-554. [PMID: 39050007 PMCID: PMC11265882 DOI: 10.55730/1300-0144.5821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/12/2024] [Accepted: 01/11/2024] [Indexed: 07/27/2024] Open
Abstract
Background/aim Type 2 diabetes mellitus (T2DM) is closely associated with atherosclerotic cardiovascular diseases (ASCVD). The objective of this study was to describe the degree of ASCVD risk factor control and their association with carotid intima-media thickness (CIMT) in T2DM patients followed up at a diabetes clinic in Southern, Sri Lanka. Materials and methods A crosssectional study was conducted to examine the association between CIMT and nonalcoholic fatty liver disease (NAFLD)in 300 T2DM patients. Both CIMT and its associations with modifiable cardiometabolic risk factors were examined using ultrasonography. The recommended optimal targets for risk factors were defined as glycated hemoglobin (HbA1C) < 7 %, absence of NAFLD, albumin-to-creatinine ratio (ACR) < 30 mg, triglyceride (TG) < 150 mg/dL, low-density lipoprotein cholesterol (LDL-C) < 100 mg/dL, high-density lipoprotein cholesterol (HDL-C) in men > 40 and in women > 50 mg/dL, systolic blood pressure (SBP) < 130 mmHg, and diastolic blood pressure (DBP) < 80 mmHg. Results SBP, DBP, LDL-C, TG, HDL-C, HbA1C, and ACR were optimally controlled in 59.3%, 75.0%, 46.7%, 84.3%, 46.0%, 33.0%, and 18.7% of patients, respectively. Notably, nearly half of the study subjects did not have NAFLD. Only three patients (1%) had achieved all therapeutic targets. There were statistically significant differences in CIMT between optimally controlled TG and suboptimally controlled TG group (p = 0.027) and between the groups with and without NAFLD (p = 0.045) when adjusted for age and duration of diabetes. CIMT showed significant and positive associations with LDL-C (p = 0.024), TG (p = 0.026), and NAFLD (p = 0.005). Among these, the presence of NAFLD had the highest odds of having higher CIMT when compared to LDL-C and TG. Conclusion The majority of patients have not achieved the recommended targets for ASCVD risk factors and are at high risk of ASCVD. It is therefore necessary to identify the reasons for not achieving the treatment targets in order to reduce the ASCVD burden by controlling LDL-C, TG, and NAFLD.
Collapse
Affiliation(s)
| | - Sarath LEKAMWASAM
- Department of Medicine, Faculty of Medicine, University of Ruhuna, Galle,
Sri Lanka
| | - Iroshani KODIKARA
- Department of Anatomy, Faculty of Medicine, University of Ruhuna, Galle,
Sri Lanka
| | | | - Lakmal FONSEKA
- Department of Medicine, Faculty of Medicine, University of Ruhuna, Galle,
Sri Lanka
| |
Collapse
|
11
|
Tan J, Fang Y, Yang C, Tay J, Tan N, Krishnan NDB, Chua BL, Zhao Y, Chen Y, Hedrick JL, Yang YY. pH-Responsive Polymeric Micelle Dynamic Complexes for Selective Killing of Helicobacter pylori. Biomacromolecules 2023; 24:5551-5562. [PMID: 37828909 DOI: 10.1021/acs.biomac.2c01374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Helicobacter pylori, the world's most common chronic infection-causing pathogen, is responsible for causing gastric ulcers, the fourth-leading cause of cancer-related death globally in 2020. In recent years, the effectiveness of the current treatment regimen (two antibiotics and one proton pump inhibitor) has often been plagued with problems such as resistance and the undesired elimination of commensal bacteria. Herein, we report the synthesis of block and random copolycarbonates, functionalized with cationic guanidinium and anionic acetate functional groups, aimed at selectively killing H. pylori in the acidic environment of the stomach, while remaining nontoxic to the commensal bacteria in the gut. The compositions of the polymers were fine-tuned so that the polymers were readily dispersed in water without any difficulty at both pH 3.0 and 7.4. The self-assembly behavior of the polymers at different pH values by dynamic light scattering showed that the random and block copolymers formed stable micelles in a simulated gastric environment (pH 3.0) while aggregated at pH 7.4. Both polymers demonstrated stronger antibacterial activity against H. pylori than the guanidinium-functionalized homopolymer without any acetate functional group at pH 3.0. The block copolymer was significantly more bactericidal at pH 3.0 across the concentrations tested, as compared to the random copolymer, while it did not show significant toxicity toward rat red blood cells (rRBCs) and HK-2 cells or bactericidal effect toward E. coli (a common gut bacterium) and nor caused aggregation of rRBCs at its effective concentration and at physiological pH of 7.4. Additionally, both the block and random copolymers were much more stable against hydrolysis at pH 3.0 than at pH 7.4. This study provides insight into the influence of both polymer architecture and dynamic assembly on the bioactivities of antimicrobial polymers, where the disassembly of coacervates into narrowly dispersed micelles at pH 3 make them potent antimicrobials aided by the protonated carboxylic acid block.
Collapse
Affiliation(s)
- Jason Tan
- Singapore Institute of Food and Biotechnology Innovation, Agency for Science Technology and Research (A*STAR), 31 Biopolis Way, Nanos #02-01, Singapore 138669, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yunhui Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 31003, China
| | - Chuan Yang
- Bioprocessing Technology Institute, Agency for Science Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Joyce Tay
- Bioprocessing Technology Institute, Agency for Science Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Nathanael Tan
- Institute of Bioengineering and Bioimaging, Agency for Science Technology and Research (A*STAR), 31 Biopolis Way, Nanos #07-01, Singapore 138669, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nithiyaa D/O Bala Krishnan
- Bioprocessing Technology Institute, Agency for Science Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Boon Lin Chua
- Bioprocessing Technology Institute, Agency for Science Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 31003, China
| | - James L Hedrick
- IBM Almaden Research Center, San Jose, California 95120, United States
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore
| |
Collapse
|
12
|
Liu S, He Y, Zhang Y, Zhang Z, Huang K, Deng L, Liao B, Zhong Y, Feng J. Targeting gut microbiota in aging-related cardiovascular dysfunction: focus on the mechanisms. Gut Microbes 2023; 15:2290331. [PMID: 38073096 PMCID: PMC10730151 DOI: 10.1080/19490976.2023.2290331] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
The global population is aging and age-related cardiovascular disease is increasing. Even after controlling for cardiovascular risk factors, readmission and mortality rates remain high. In recent years, more and more in-depth studies have found that the composition of the gut microbiota and its metabolites, such as trimethylamine N-oxide (TMAO), bile acids (BAs), and short-chain fatty acids (SCFAs), affect the occurrence and development of age-related cardiovascular diseases through a variety of molecular pathways, providing a new target for therapy. In this review, we discuss the relationship between the gut microbiota and age-related cardiovascular diseases, and propose that the gut microbiota could be a new therapeutic target for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yali Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Zhaolun Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Keming Huang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
13
|
Bestavashvili A, Glazachev O, Ibragimova S, Suvorov A, Bestavasvili A, Ibraimov S, Zhang X, Zhang Y, Pavlov C, Syrkina E, Syrkin A, Kopylov P. Impact of Hypoxia-Hyperoxia Exposures on Cardiometabolic Risk Factors and TMAO Levels in Patients with Metabolic Syndrome. Int J Mol Sci 2023; 24:14498. [PMID: 37833946 PMCID: PMC10572339 DOI: 10.3390/ijms241914498] [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: 07/04/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
Along with the known risk factors of cardiovascular diseases (CVDs) constituting metabolic syndrome (MS), the gut microbiome and some of its metabolites, in particular trimethylamine-N-oxide (TMAO), are actively discussed. A prolonged stay under natural hypoxic conditions significantly and multi-directionally changes the ratio of gut microbiome strains and their metabolites in feces and blood, which is the basis for using hypoxia preconditioning for targeted effects on potential risk factors of CVD. A prospective randomized study included 65 patients (32 females) with MS and optimal medical therapy. Thirty-three patients underwent a course of 15 intermittent hypoxic-hyperoxic exposures (IHHE group). The other 32 patients underwent sham procedures (placebo group). Before and after the IHHE course, patients underwent liver elastometry, biochemical blood tests, and blood and fecal sampling for TMAO analysis (tandem mass spectrometry). No significant dynamics of TMAO were detected in both the IHHE and sham groups. In the subgroup of IHHE patients with baseline TMAO values above the reference (TMAO ≥ 5 μmol/l), there was a significant reduction in TMAO plasma levels. But the degree of reduction in total cholesterol (TCh), low-density lipoprotein (LDL), and regression of liver steatosis index was more pronounced in patients with initially normal TMAO values. Despite significant interindividual variations, in the subgroup of IHHE patients with MS and high baseline TMAO values, there were more significant reductions in cardiometabolic and hepatic indicators of MS than in controls. More research is needed to objectify the prognostic role of TMAO and the possibilities of its correction using hypoxia adaptation techniques.
Collapse
Affiliation(s)
- Afina Bestavashvili
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Oleg Glazachev
- Department of Normal Physiology, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Shabnam Ibragimova
- Department of Therapy of the Institute of Professional Education, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Alexander Suvorov
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | - Shevket Ibraimov
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Xinliang Zhang
- Department of Normal Physiology, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Yong Zhang
- The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology, Harbin Medical University, Harbin 150081, China
| | - Chavdar Pavlov
- Department of Therapy of the Institute of Professional Education, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Department of Gastroenterology, Botkin Hospital, 125284 Moscow, Russia
| | - Elena Syrkina
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Abram Syrkin
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Philipp Kopylov
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
14
|
Ciccone MM, Lepera ME, Guaricci AI, Forleo C, Cafiero C, Colella M, Palmirotta R, Santacroce L. Might Gut Microbiota Be a Target for a Personalized Therapeutic Approach in Patients Affected by Atherosclerosis Disease? J Pers Med 2023; 13:1360. [PMID: 37763128 PMCID: PMC10532785 DOI: 10.3390/jpm13091360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
In recent years, the increasing number of studies on the relationship between the gut microbiota and atherosclerosis have led to significant interest in this subject. The gut microbiota, its metabolites (metabolome), such as TMAO, and gut dysbiosis play an important role in the development of atherosclerosis. Furthermore, inflammation, originating from the intestinal tract, adds yet another mechanism by which the human ecosystem is disrupted, resulting in the manifestation of metabolic diseases and, by extension, cardiovascular diseases. The scientific community must understand and elucidate these mechanisms in depth, to gain a better understanding of the relationship between atherosclerosis and the gut microbiome and to promote the development of new therapeutic targets in the coming years. This review aims to present the knowledge acquired so far, to trigger others to further investigate this intriguing topic.
Collapse
Affiliation(s)
- Marco Matteo Ciccone
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Mario Erminio Lepera
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Andrea Igoren Guaricci
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Cinzia Forleo
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Concetta Cafiero
- Area of Molecular Pathology, Anatomic Pathology Unit, Fabrizio Spaziani Hospital, 03100 Frosinone, Italy;
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| | - Raffele Palmirotta
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| |
Collapse
|
15
|
Guiducci L, Nicolini G, Forini F. Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites 2023; 13:760. [PMID: 37367916 DOI: 10.3390/metabo13060760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
The cardiovascular and metabolic disorders, collectively known as cardiometabolic disease (CMD), are high morbidity and mortality pathologies associated with lower quality of life and increasing health-care costs. The influence of the gut microbiota (GM) in dictating the interpersonal variability in CMD susceptibility, progression and treatment response is beginning to be deciphered, as is the mutualistic relation established between the GM and diet. In particular, dietary factors emerge as pivotal determinants shaping the architecture and function of resident microorganisms in the human gut. In turn, intestinal microbes influence the absorption, metabolism, and storage of ingested nutrients, with potentially profound effects on host physiology. Herein, we present an updated overview on major effects of dietary components on the GM, highlighting the beneficial and detrimental consequences of diet-microbiota crosstalk in the setting of CMD. We also discuss the promises and challenges of integrating microbiome data in dietary planning aimed at restraining CMD onset and progression with a more personalized nutritional approach.
Collapse
Affiliation(s)
- Letizia Guiducci
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| | | | - Francesca Forini
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
16
|
Han S, Cai L, Chen P, Kuang W. A study of the correlation between stroke and gut microbiota over the last 20years: a bibliometric analysis. Front Microbiol 2023; 14:1191758. [PMID: 37350780 PMCID: PMC10282156 DOI: 10.3389/fmicb.2023.1191758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Purpose This study intends to uncover a more thorough knowledge structure, research hotspots, and future trends in the field by presenting an overview of the relationship between stroke and gut microbiota in the past two decades. Method Studies on stroke and gut microbiota correlations published between 1st January 2002 and 31st December 2021 were retrieved from the Web of Science Core Collection and then visualized and scientometrically analyzed using CiteSpace V. Results A total of 660 papers were included in the study, among which the United States, the United Kingdom, and Germany were the leading research centers. Cleveland Clinic, Southern Medical University, and Chinese Academy of Science were the top three institutions. The NATURE was the most frequently co-cited journal. STANLEY L HAZEN was the most published author, and Tang WHW was the most cited one. The co-occurrence analysis revealed eight clusters (i.e., brain-gut microbiota axis, fecal microbiome transplantation, gut microbiota, hypertension, TMAO, ischemic stroke, neuroinflammation, atopobiosis). "gut microbiota," "Escherichia coli," "cardiovascular disease," "risk," "disease," "ischemic stroke," "stroke," "metabolism," "inflammation," and "phosphatidylcholine" were the most recent keyword explosions. Conclusion Findings suggest that in the next 10 years, the number of publications produced annually may increase significantly. Future research trends tend to concentrate on the mechanisms of stroke and gut microbiota, with the inflammation and immunological mechanisms, TMAO, and fecal transplantation as hotspots. And the relationship between these mechanisms and a particular cardiovascular illness may also be a future research trend.
Collapse
Affiliation(s)
- Shengnan Han
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Longhui Cai
- First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peipei Chen
- School of Medical Technology, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
17
|
Wang L, Lei J, Wang R, Li K. Non-Traditional Risk Factors as Contributors to Cardiovascular Disease. Rev Cardiovasc Med 2023; 24:134. [PMID: 39076735 PMCID: PMC11273054 DOI: 10.31083/j.rcm2405134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/18/2022] [Accepted: 01/11/2023] [Indexed: 07/31/2024] Open
Abstract
Cardiovascular disease (CVD) remains one of the primary causes of morbidity and mortality worldwide. Classic cardiovascular risk factors, such as hypertension, diabetes mellitus (DM), hyperlipidemia, and smoking, have been well identified and given increased attention in clinical practice. However, the incidence and prevalence of CVD remains high, especially in developing countries. Therefore, there has been more attention to non-traditional CVD risk factors such as gut microbiota, sleep disorders, dietary structure, and psychosocial factors in their important roles in the development of CVD. In this review we summarize the association of non-traditional risk factors with CVD with the aim of further reducing the risk of CVD.
Collapse
Affiliation(s)
- Lina Wang
- Department of Cardiology, Hebei Yanda Hospital, 065201 Langfang, Hebei, China
| | - Jingshu Lei
- Department of Cardiology, Hebei Yanda Hospital, 065201 Langfang, Hebei, China
| | - Ruiying Wang
- Department of Hematology, Hebei Yanda Lu Daopei Hospital, 065201 Langfang, Hebei, China
| | - Kuibao Li
- Department of Cardiology, Hebei Yanda Hospital, 065201 Langfang, Hebei, China
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, 100016 Beijing, China
| |
Collapse
|
18
|
Hypotheses on Atherogenesis Triggering: Does the Infectious Nature of Atherosclerosis Development Have a Substruction? Cells 2023; 12:cells12050707. [PMID: 36899843 PMCID: PMC10001176 DOI: 10.3390/cells12050707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Since the end of the 20th century, it has been clear that atherosclerosis is an inflammatory disease. However, the main triggering mechanism of the inflammatory process in the vascular walls is still unclear. To date, many different hypotheses have been put forward to explain the causes of atherogenesis, and all of them are supported by strong evidence. Among the main causes of atherosclerosis, which underlies these hypotheses, the following can be mentioned: lipoprotein modification, oxidative transformation, shear stress, endothelial dysfunction, free radicals' action, homocysteinemia, diabetes mellitus, and decreased nitric oxide level. One of the latest hypotheses concerns the infectious nature of atherogenesis. The currently available data indicate that pathogen-associated molecular patterns from bacteria or viruses may be an etiological factor in atherosclerosis. This paper is devoted to the analysis of existing hypotheses for atherogenesis triggering, and special attention is paid to the contribution of bacterial and viral infections to the pathogenesis of atherosclerosis and cardiovascular disease.
Collapse
|
19
|
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
|
20
|
Chi M, Jiang T, He X, Peng H, Li Y, Zhang J, Wang L, Nian Q, Ma K, Liu C. Role of Gut Microbiota and Oxidative Stress in the Progression of Transplant-Related Complications following Hematopoietic Stem Cell Transplantation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3532756. [PMID: 37113743 PMCID: PMC10129428 DOI: 10.1155/2023/3532756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/12/2022] [Accepted: 11/24/2022] [Indexed: 04/29/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT), also known as bone marrow transplantation, has curative potential for various hematologic malignancies but is associated with risks such as graft-versus-host disease (GvHD), severe bloodstream infection, viral pneumonia, idiopathic pneumonia syndrome (IPS), lung fibrosis, and sinusoidal obstruction syndrome (SOS), which severely deteriorate clinical outcomes and limit the wide application of HSCT. Recent research has provided important insights into the effects of gut microbiota and oxidative stress (OS) on HSCT complications. Therefore, based on recent studies, we describe intestinal dysbiosis and OS in patients with HSCT and review recent molecular findings underlying the causal relationships of gut microbiota, OS, and transplant-related complications, focusing particularly on the involvement of gut microbiota-mediated OS in postengraftment complications. Also, we discuss the use of antioxidative and anti-inflammatory probiotics to manipulate gut microbiota and OS, which have been associated with promising effects in improving HSCT outcomes.
Collapse
Affiliation(s)
- Mingxuan Chi
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Tao Jiang
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
- Department of Hematology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province 610072, China
| | - Xing He
- School of Clinical Medicine, Chengdu Medical College, China
| | - Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunlong Li
- Department of Urology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiong Zhang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Li Wang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Qing Nian
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
- Department of Blood Transfusion, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chi Liu
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Renal Disease Clinical Research Center, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| |
Collapse
|
21
|
Cheng Q, Fan C, Liu F, Li Y, Hou H, Ma Y, Tan Y, Li Y, Hai Y, Wu T, Zhang L, Zhang Y. Structural and functional dysbiosis of gut microbiota in Tibetan subjects with coronary heart disease. Genomics 2022; 114:110483. [PMID: 36115504 DOI: 10.1016/j.ygeno.2022.110483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 08/24/2022] [Accepted: 09/13/2022] [Indexed: 01/14/2023]
Abstract
The gut microbiota plays a crucial role in coronary heart disease (CHD). However, only a few studies focusing on the relationship between gut microbiota and CHD in ethnic populations are available. Here, we employed shotgun sequencing of the gut metagenome to analyze the taxonomic composition and functional annotation of the gut microbiota of 14 CHD patients, 13 patients with non-stenosis coronary heart disease (NCHD), and 18 healthy controls (HT) in Tibetan subjects. We found that the α-diversity of the gut microbiota was not significantly different among the three groups., whereas β-diversity was significantly altered in the CHD group compared with HT. Based on the receiver operating characteristic curve (ROC) analysis, the relative abundance of Proteobacteria species effectively distinguished patients with CHD from the control group. Most of the enriched species belonged to Proteobacteria. The pathways that contributed the most to the differences between groups were amino acid metabolism-related pathways, especially lysine biosynthesis. The enzymes of the lysine biosynthesis pathway, including K01714 and K00821, were significantly decreased in the CHD group. Our findings increase the understanding of the association between CHD pathogenesis and gut microbiota in the Tibetan population, thus paving the way for the development of improved diagnostic methods and treatments for Tibetan patients with CHD.
Collapse
Affiliation(s)
- Qi Cheng
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Fan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengyun Liu
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Yuan Li
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Haiwen Hou
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Yan Ma
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Yueqing Tan
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Yuxian Li
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Yue Hai
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China
| | - Tianyi Wu
- National Key Laboratory of High Altitude Medicine, Qinghai High Altitude Medical Research Institute, Xining 810012, China; Qinghai Province Cardiovascular and Cerebrovascular Disease Specialist Hospital, Xining 810012, China.
| | - Liangzhi Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
| | - Yanming Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
| |
Collapse
|
22
|
Chiang EPI, Syu JN, Hung HC, Rodriguez RL, Wang WJ, Chiang ER, Chiu SC, Chao CY, Tang FY. N-3 polyunsaturated fatty acids block the trimethylamine-N-oxide- ACE2- TMPRSS2 cascade to inhibit the infection of human endothelial progenitor cells by SARS-CoV-2. J Nutr Biochem 2022; 109:109102. [PMID: 35817244 PMCID: PMC9264727 DOI: 10.1016/j.jnutbio.2022.109102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2(SARS-CoV-2) is a novel coronavirus that infects many types of cells and causes cytokine storms, excessive inflammation, acute respiratory distress to induce failure of respiratory system and other critical organs. In this study, our results showed that trimethylamine-N-oxide (TMAO), a metabolite generated by gut microbiota, acts as a regulatory mediator to enhance the inerleukin-6 (IL-6) cytokine production and the infection of human endothelial progenitor cells (hEPCs) by SARS-CoV-2. Treatment of N-3 polyunsaturated fatty acids (PUFAs) such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) could effectively block the entry of SARS-CoV-2 in hEPCs. The anti-infection effects of N-3 PUFAs were associated with the inactivation of NF-κB signaling pathway, a decreased expression of the entry receptor angiotensin-converting enzyme 2 (ACE2) and downstream transmembrane serine protease 2 in hEPCs upon the stimulation of TMAO. Treatment of DHA and EPA further effectively inhibited TMAO-mediated expression of IL-6 protein, probably through an inactivation of MAPK/p38/JNK signaling cascades and a downregulation of microRNA (miR)-221 in hEPCs. In conclusion, N-3 PUFAs such as DHA and EPA could effectively act as preventive agents to block the infection of SARS-CoV-2 and IL-6 cytokine production in hEPCs upon the stimulation of TMAO.
Collapse
Affiliation(s)
- En-Pei Isabel Chiang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan, Republic of China; Innovation and Development Center of Sustainable Agriculture, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Jia-Ning Syu
- Biomedical Science Laboratory, Department of Nutrition, China Medical University, Taichung, Taiwan, Republic of China
| | - Hung-Chang Hung
- Department of Internal Medicine, Nantou Hospital, Ministry of Health and Welfare, Nantou City, Taiwan, Republic of China
| | - Raymond L Rodriguez
- Department of Molecular and Cellular Biology, University of California, Davis, California, USA
| | - Wei-Jan Wang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, Republic of China
| | - En-Rung Chiang
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China; National Yang Ming Chiao Tung University, School of Medicine, Taipei, Taiwan, Republic of China
| | - Shao-Chih Chiu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, Republic of China; Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Che-Yi Chao
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan, Republic of China; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, Republic of China
| | - Feng-Yao Tang
- Biomedical Science Laboratory, Department of Nutrition, China Medical University, Taichung, Taiwan, Republic of China.
| |
Collapse
|
23
|
Long D, Mao C, Zhang X, Liu Y, Shangguan X, Zou M, Zhu Y, Wang X. Coronary heart disease and gut microbiota: A bibliometric and visual analysis from 2002 to 2022. Front Cardiovasc Med 2022; 9:949859. [PMID: 36158832 PMCID: PMC9493042 DOI: 10.3389/fcvm.2022.949859] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/15/2022] [Indexed: 12/03/2022] Open
Abstract
Background Existing studies have indicated that gut microbiota is closely related to the occurrence and development of coronary heart disease(CHD). Gut microbiota and its metabolites may be important diagnostic markers for CHD in the future and are expected to become new targets for the prevention and treatment of CHD. However, the current studies exploring the link between CHD and gut microbiota are miscellaneous and poorly targeted, without bibliometric analysis available. Objective The purpose of this research was to perform a bibliometric and visual analysis of published papers on the relationship between CHD and gut microbiota. The study also sought to identify principal authors, institutions, and countries to analyze the research status and trends of gut microbiota research in the field of CHD. Methods The Web of Science Core Collection (WoSCC) database was searched for publications on CHD and gut microbiota between 2002 and 2022. CiteSpace 5.8. R1, VOSviewer 1.6.16, and Microsoft Excel 2019 software tools were utilized to perform this bibliometric analysis and visualization. Results There were 457 qualified publications found in total, with the annual number of publications increasing. The United States dominated in this field. Hazen, Stanley l was the author of the most papers. Cleveland Clinic published the most papers of any institution. The six main clusters’ specific characteristics were discovered through analysis of the co-occurrence of keywords: inflammation, diet, trimethylamine n-oxide, metabolism, cardiovascular disease, and myocardial infarction. Newly emerging research has focused predominantly on gut microbiota metabolites and recent strategies for intervention in coronary atherosclerosis. Conclusion These results provided a useful perspective on current research and future prospects for the research on the link between CHD and gut microbiota, which may help researchers to select suitable collaborators and facilitate their research to elucidate the underlying molecular mechanisms of CHD, including the causes, prevention, and treatment.
Collapse
Affiliation(s)
- Dan Long
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Chenhan Mao
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyue Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yaxuan Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xueli Shangguan
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Menglong Zou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Ying Zhu,
| | - Xindong Wang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Xindong Wang,
| |
Collapse
|
24
|
Ghorbani Z, Kazemi A, Bartolomaeus TUP, Martami F, Noormohammadi M, Salari A, Löber U, Balou HA, Forslund SK, Mahdavi-Roshan M. The effect of probiotic and synbiotic supplementation on lipid parameters among patients with cardiometabolic risk factors: a systematic review and meta-analysis of clinical trials. Cardiovasc Res 2022; 119:933-956. [PMID: 35934838 DOI: 10.1093/cvr/cvac128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
Although the available evidence emphasizes the beneficial effects of probiotics in normalizing various cardiometabolic markers, there is still substantial uncertainty in this regard. Thus, we set out to determine the effect sizes of probiotics on blood lipid parameters more coherently. A systematic literature search of the Medline (PubMed) and Scopus databases was conducted from inception to February 12, 2021, applying both MeSH terms and free text terms to find the relevant randomized controlled trials (RCTs). The meta-analysis was conducted based on a random-effect model to calculate the mean effect sizes demonstrated as weighted mean differences (WMD) and the 95% confidence intervals (95%CI). To explore the heterogeneity, the Cochrane Chi-squared test, and analysis of Galbraith plots were performed. Meta-analysis of data from 40 RCTs (n = 2795) indicated a significant decrease in serum/plasma triglyceride (WMD (95%CI) -12.26 (-17.11- -7.41) mg/dL; P-value <0.001; I2 (%)= 29.9; P heterogeneity = 0.034)), total cholesterol (with high heterogeneity) (WMD (95%CI) -8.43 (-11.90- -4.95) mg/dL; P-value <0.001; I2 (%) =56.8; P heterogeneity < 0.001), LDL-C (WMD (95%CI) -5.08 (-7.61, -2.56) mg/dL; P-value <0.001; I2 (%) =42.7; P heterogeneity =0.002), and HDL-C (with high heterogeneity) (WMD (95%CI) 1.14 (0.23, 2.05) mg/dL; P-value =0.014; I2 (%) = 59.8; P heterogeneity < 0.001) following receiving probiotic/synbiotic supplements. Collectively, the current preliminary evidence supports the effectiveness of probiotics/synbiotics in improving dyslipidemia and various lipid parameters more prominently among subjects with hyperlipidemia, diabetes, and metabolic syndrome. However, large and well conducted RCTs are required to provide further convincing support for these results.
Collapse
Affiliation(s)
- Zeinab Ghorbani
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Asma Kazemi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Theda U P Bartolomaeus
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany
| | - Fahimeh Martami
- School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Morvarid Noormohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ulrike Löber
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany
| | - Heydar Ali Balou
- Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sofia K Forslund
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Berlin, Germany.,Structural and Computational Biology Unit, European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Marjan Mahdavi-Roshan
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Clinical Nutrition, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
25
|
Dong Y, Wu X, Han L, Bian J, He C, El-Omar E, Gong L, Wang M. The Potential Roles of Dietary Anthocyanins in Inhibiting Vascular Endothelial Cell Senescence and Preventing Cardiovascular Diseases. Nutrients 2022; 14:nu14142836. [PMID: 35889793 PMCID: PMC9316990 DOI: 10.3390/nu14142836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease (CVD) is a group of diseases affecting the heart and blood vessels and is the leading cause of morbidity and mortality worldwide. Increasingly more evidence has shown that the senescence of vascular endothelial cells is the key to endothelial dysfunction and cardiovascular diseases. Anthocyanin is a type of water-soluble polyphenol pigment and secondary metabolite of plant-based food widely existing in fruits and vegetables. The gut microbiome is involved in the metabolism of anthocyanins and mediates the biological activities of anthocyanins and their metabolites, while anthocyanins also regulate the growth of specific bacteria in the microbiota and promote the proliferation of healthy anaerobic flora. Accumulating studies have shown that anthocyanins have antioxidant, anti-inflammatory, and anti-aging effects. Many animal and in vitro experiments have also proven that anthocyanins have protective effects on cardiovascular-disease-related dysfunction. However, the molecular mechanism of anthocyanin in eliminating aging endothelial cells and preventing cardiovascular diseases is very complex and is not fully understood. In this systematic review, we summarize the metabolism and activities of anthocyanins, as well as their effects on scavenging senescent cells and cardioprotection.
Collapse
Affiliation(s)
- Yonghui Dong
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (Y.D.); (X.W.); (L.H.); (C.H.)
| | - Xue Wu
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (Y.D.); (X.W.); (L.H.); (C.H.)
| | - Lin Han
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (Y.D.); (X.W.); (L.H.); (C.H.)
| | - Ji Bian
- Kolling Institute, Sydney Medical School, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW 2065, Australia;
| | - Caian He
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (Y.D.); (X.W.); (L.H.); (C.H.)
| | - Emad El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Lan Gong
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, NSW 2052, Australia;
- Correspondence: (L.G.); (M.W.)
| | - Min Wang
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (Y.D.); (X.W.); (L.H.); (C.H.)
- Correspondence: (L.G.); (M.W.)
| |
Collapse
|
26
|
The Effects of Probiotics on Inflammation, Endothelial Dysfunction, and Atherosclerosis Progression: A Mechanistic Overview. Heart Lung Circ 2022; 31:e45-e71. [PMID: 35153150 DOI: 10.1016/j.hlc.2021.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/07/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The relationship between the intestinal microbiota dysbiosis, inflammation, and cardiovascular disorders (CVDs) has become evident, based on a growing body of literature from animal models and human studies. On the other hand, probiotics are believed to have promising effects on modifying dysbiosis and protecting against CVDs. OBJECTIVE This narrative review provides an overview of the link between gut microbiota, inflammation, endothelial dysfunction, and atherosclerosis. The influences of probiotic supplementation on biomarkers contributing to these conditions as the primary underlying risk factors for developing CVDs are also discussed. METHODS An up-to-date review was performed of the available evidence from experimental studies, clinical trials, and meta-analyses, considering their challenges and limitations. It also aimed to provide mechanistic insight into the likely mechanisms of probiotics that could prevent atherosclerosis initiation and progression. RESULTS Probiotic supplementation seems to be associated with reduced levels of inflammation and oxidative stress biomarkers (C-reactive protein, tumour necrosis factor-α, interleukin (IL)-6, IL-12, and malondialdehyde). Further, these agents might enhance antioxidant factors (IL-10, total antioxidant status, total antioxidant capacity, glutathione, and nitric oxide). Probiotics also appear to improve intestinal barrier integrity, reduce leakage of harmful metabolites (e.g., lipopolysaccharides), inhibit pro-inflammatory signalling pathways, and possibly suppress the formation of trimethylamine/trimethylamine oxide. Probiotics have also been found to enhance endothelial function and halter thrombosis. CONCLUSION The current clinical evidence underlines belief that probiotics might be associated with reduced levels of inflammation biomarkers. Experimental evidence reports that the beneficial effects of probiotics seem to be mainly imposed by triggering the secretion of short-chain fatty acids and bile acids, in addition to suppressing the NF-κB signalling pathway. However, the current studies are still in their infancy and it is of high priority to design further research on the topic.
Collapse
|
27
|
Tam I, Liu PH, Ma W, Cao Y, Jovani M, Wu K, Rimm EB, Strate LL, Giovannucci EL, Chan AT. History of Diverticulitis and Risk of Incident Cardiovascular Disease in Men: A Cohort Study. Dig Dis Sci 2022; 67:1337-1344. [PMID: 33770332 PMCID: PMC8464617 DOI: 10.1007/s10620-021-06949-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 03/08/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Diverticulitis and cardiovascular disease (CVD) are two highly prevalent disorders sharing common risk factors which are hypothesized to have an inflammatory basis. AIMS To examine the association between history of diverticulitis and risk of incident CVD. METHODS We conducted a prospective cohort study of 43,904 men aged 40 to 75 years without a history of CVD (fatal or nonfatal myocardial infarction and stroke) at enrollment who were followed up from 1986 to 2012 in the Health Professionals Follow-Up Study. Lifestyle factors, dietary intake, and disease information were self-reported biennially or quadrennially. Incident diverticulitis and CVD were confirmed by review of medical records. We used Cox proportional hazard models to calculate age- and multivariable-adjusted hazard ratios (HR) and 95% confidence intervals (CI) of incident CVD. We conducted a stratified analysis according to the presence or absence of CVD risk factors (smoking, hypertension, hyperlipidemia, and diabetes). RESULTS We identified 3848 incident cases of CVD during 856,319 person-years of follow-up. Men with diverticulitis had higher incidence of CVD (727 cases per 100,000 person-years) compared to men without diverticulitis [446 cases per 100,000 person-years, multivariate HR of 1.35 (95% CI 1.07-1.70)]. The association of diverticulitis and subsequent CVD appeared more evident among men without known CVD risk factors (HR 4.06, 95% CI 2.04-8.08) compared to those with one or more CVD risk factors (HR 1.27, 95% CI 0.98-1.63). CONCLUSIONS Diverticulitis may be an independent risk factor of incident CVD, suggesting possible common etiopathogenic mechanisms. Diagnosis of diverticulitis underscores the importance of preventive measures to reduce future CVD.
Collapse
Affiliation(s)
- Idy Tam
- Tufts University School of Medicine, Boston, MA, USA
| | - Po-Hong Liu
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
- Digestive and Liver Diseases Division, UT Southwestern Medical Center, Dallas, TX, USA
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yin Cao
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Manol Jovani
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lisa L Strate
- Division of Gastroenterology, University of Washington School of Medicine, Harborview Medical Center, Seattle, WA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
28
|
Liu X, Shao Y, Tu J, Sun J, Li L, Tao J, Chen J. Trimethylamine-N-oxide-stimulated hepatocyte-derived exosomes promote inflammation and endothelial dysfunction through nuclear factor-kappa B signaling. ANNALS OF TRANSLATIONAL MEDICINE 2022; 9:1670. [PMID: 34988179 PMCID: PMC8667148 DOI: 10.21037/atm-21-5043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022]
Abstract
Background Trimethylamine-N-oxide (TMAO) has been proven to be a new proatherogenic compound for promoting inflammation and endothelial dysfunction. Hepatocyte-derived exosomes (Exos), including those derived from hepatocytes, play a pivotal role in the regulation of inflammation and endothelial function. As TMAO is produced in the liver, hepatocytes may be the potential target of TMAO. However, it is not yet clear whether TMAO can directly stimulate hepatocytes to produce Exos to mediate the detrimental effects of TMAO on vascular endothelial cells (VECs). Methods Hepatocytes treated with TMAO and Exos (TMAO-Exos) were isolated from the supernatant, and added to human aortic endothelial cells (HAECs). The expressions of interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor-α (TNF-α) were detected by quantitative polymerase chain reaction (qPCR). Cell apoptosis was evaluated using Hoechst 33342 staining and flow cytometry assay, and cell migration was assessed by scratch and transwell assay. C57BL/6 mice were treated with Exos for 24 h and the thoracic aortas were isolated, then the in vitro aortic ring bioassay was conducted to determine the changes of vasodilation. The expressions of cluster of differentiation 81, tumor susceptibility gene 101, nuclear factor-kappa B (NF-κB) p65, and Phospho-NF-κB p65 were detected by western blotting. The micro ribonucleic acid (miRNA) profiles of the Exos were then identified using RNA-sequencing and validated by qPCR. The miRNA-messenger RNA networks were constructed, and the biological functions of the target genes were annotated using bioinformatics methods. Results TMAO was found to stimulate hepatocytes to release Exos that could be taken up by HAECs, thus inducing inflammation and cell apoptosis, impairing cell migration, and inhibiting endothelium-dependent vasodilation. Additionally, the miRNAs such as miR-302d-3p carried by the TMAO-Exos were quite different to those in the TMAO-free group. A further analysis showed that the potential target genes for these miRNAs, such as mitogen-activated protein kinase 8, caspase 9 and BCL2-like 11, appeared to be involved with inflammation and endothelial function. Finally, we found that NF-κB signaling could be activated by TMAO-Exos. Conclusions These novel findings provide evidence that TMAO can indirectly talk to VECs by promoting hepatocytes to produce Exos that carry important genetic information.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Yijia Shao
- Department of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Jiazichao Tu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Jiapan Sun
- Department of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Lifu Li
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Jun Tao
- Department of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Jimei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| |
Collapse
|
29
|
El-Shal LM, El-Star AAA, Azmy AM, Elnegris HM. The possible protective role of N-acetyl cysteine on duodenal mucosa of high fat diet and orlistat treated adult male albino rats and the active role of tumor necrosis factor α (TNFα) and Interleukin 6 (IL6) (histological and biochemical study). Ultrastruct Pathol 2022; 46:18-36. [PMID: 34979873 DOI: 10.1080/01913123.2021.2007194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Obesity is a major universal health issue linked to a majority of illness. AIM To evaluate the histological and biochemical changes occurred in the duodenal mucosa of high fat diet HFD and orlistat fed rats and to assess the possible protective role of N-acetyl cysteine NAC supplementation. MATERIAL AND METHOD Sixty male albino rats weighing 180-200 g were classified randomly into control group I and three experimental groups (HFD group II, HFD + orlistat group III, and HFD + orlistat + NAC group IV). All experimental groups received HFD alone/and treatment for 6 weeks. Group III received orlistat (32 mg/kg/day) before meals and group IV received the same regimen as group III in addition to NAC (230 mg/kg/day) after meals. After completion of the experiment, duodenal sections were processed for histological examination, oxidative stress parameters, and semiqualitative real time PCR for proinflammatory mediators TNFα and IL6 evaluation. Also, plasma lipid parameters were assessed and morphometric duodenal results were analyzed statistically. RESULTS By histological examination of HFD and (HFD + orlistat) groups, we found severe to moderate duodenal structural disturbances, increased goblet cells, collagen fibers, and BAX and iNOS immunostaining. By Biochemical examination, both groups showed increased proinflammatory markers level (TNFα and IL6) with decreased all antioxidant parameters and increased MDA. Moreover, NAC treatment in group IV significantly reduced all structural changes, levels of proinflammatory mediators and increased all antioxidant parameter levels and decreased MDA. CONCLUSION All findings elucidated that NAC could be accounted to be a useful drug for protection of duodenal mucosa of HFD and orlistat treated animals.
Collapse
Affiliation(s)
- Laila Moustafa El-Shal
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Alyaa A Abd El-Star
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abeer M Azmy
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Heba M Elnegris
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.,Department of Histology and Cell Biology, Faculty of Medicine, Badr University in Cairo, Cairo, Egypt
| |
Collapse
|
30
|
Hakhamaneshi MS, Abdolahi A, Vahabzadeh Z, Abdi M, Andalibi P. Toll-Like Receptor 4: A Macrophage Cell Surface Receptor Is Activated By Trimethylamine-N-Oxide. CELL JOURNAL 2021; 23:516-522. [PMID: 34837678 PMCID: PMC8588815 DOI: 10.22074/cellj.2021.7849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/03/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Trimethylamine-N-Oxide (TMAO) is considered as a risk factor for atherosclerosis which further leads to inflammation during atherosclerosis. The exact mechanism(s) by which TMAO induces the inflammatory reactions remains to be determined. TMAO can cause the endoplasmic reticulum (ER) stress that triggers activation of Toll-Like Receptors (TLRs). In macrophages, this process stimulates the production of proinflammatory cytokines. This study designed to evaluate the expression level of TLR4 in TMAO-treated macrophages. MATERIALS AND METHODS In this experimental study, different concentrations of TMAO (37.5, 75, 150, and 300 μM) were exposed to murine macrophage (J774A.1 cell line) for 8, 18, 24, and 48 hours. The cells were also treated with 2.5 mM of 4-phenyl butyric acid as well as 2μg/ml of tunicamycin respectively as negative and positive controls for inducing ER-stress. We measured the viability of treated cells by the MTT test. Besides, the expression levels of TLR4 gene and protein were evaluated using western blotting and reverse transcription- quantitative polymerase chain reaction (RT-qPCR) analysis. One-Way ANOVA was used for statistical analysis. RESULTS No cell death was observed in treated cells. The cells treated with 150 and 300 μM doses of TMAO for 24 hours showed a significant elevation in the protein and/or mRNA levels of TLR4 when compared to normal control or tunicamycin-treated cells. CONCLUSION Our results may in part elucidate the mechanism by which TMAO induces the macrophage inflammatory reactions in response to the induction of ER stress, similar to what happens during atherosclerosis. It also provides documentation to support the direct contribution of TLR4 in TMAO-induced inflammation.
Collapse
Affiliation(s)
| | - Alina Abdolahi
- Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Zakaria Vahabzadeh
- Department of Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran. .,Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Abdi
- Cellular and Molecular Research Centre, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Pedram Andalibi
- Department of Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
31
|
Fernández AI, Bermejo J, Yotti R, Martínez-Gonzalez MÁ, Mira A, Gophna U, Karlsson R, Al-Daccak R, Martín-Demiguel I, Gutiérrez-Ibanes E, Charron D, Fernández-Avilés F. The impact of Mediterranean diet on coronary plaque vulnerability, microvascular function, inflammation and microbiome after an acute coronary syndrome: study protocol for the MEDIMACS randomized, controlled, mechanistic clinical trial. Trials 2021; 22:795. [PMID: 34772433 PMCID: PMC8588729 DOI: 10.1186/s13063-021-05746-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background Primary prevention trials have demonstrated that the traditional Mediterranean diet is associated with a reduction in cardiovascular mortality and morbidity. However, this benefit has not been proven for secondary prevention after an acute coronary syndrome (ACS). We hypothesized that a high-intensity Mediterranean diet intervention after an ACS decreases the vulnerability of atherosclerotic plaques by complex interactions between anti-inflammatory effects, microbiota changes and modulation of gene expression. Methods The MEDIMACS project is an academically funded, prospective, randomized, controlled and mechanistic clinical trial designed to address the effects of an active randomized intervention with the Mediterranean diet on atherosclerotic plaque vulnerability, coronary endothelial dysfunction and other mechanistic endpoints. One hundred patients with ACS are randomized 1:1 to a monitored high-intensity Mediterranean diet intervention or to a standard-of-care arm. Adherence to diet is assessed in both arms using food frequency questionnaires and biomarkers of compliance. The primary endpoint is the change (from baseline to 12 months) in the thickness of the fibrous cap of a non-significant atherosclerotic plaque in a non-culprit vessel, as assessed by repeated optical coherence tomography intracoronary imaging. Indices of coronary vascular physiology and changes in gastrointestinal microbiota, immunological status and protein and metabolite profiles will be evaluated as secondary endpoints. Discussion The results of this trial will address the key effects of dietary habits on atherosclerotic risk and will provide initial data on the complex interplay of immunological, microbiome-, proteome- and metabolome-related mechanisms by which non-pharmacological factors may impact the progression of coronary atherosclerosis after an ACS. Trial registration ClinicalTrials.govNCT03842319. Registered on 13 May 2019 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05746-z.
Collapse
Affiliation(s)
- Ana I Fernández
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Madrid, Spain
| | - Javier Bermejo
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Madrid, Spain.
| | - Raquel Yotti
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Madrid, Spain
| | - Miguel Ángel Martínez-Gonzalez
- Department of Preventive Medicine and Public Health, University of Navarra, IDISNA, CIBEROBN, Pamplona, Spain.,Department of Nutrition, Harvard TH Chan School of Public Health, Boston, USA
| | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, CSISP-FISABIO, and CIBERESP, Valencia, Spain
| | - Uri Gophna
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Roger Karlsson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy of the University of Gothenburg; Sweden Nanoxis Consulting AB; Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Reem Al-Daccak
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-97f, Université Paris-Diderot, HLA et Médecine, Labex Transplantex, Hôpital Saint-Louis, Paris, France
| | - Irene Martín-Demiguel
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Madrid, Spain
| | - Enrique Gutiérrez-Ibanes
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Madrid, Spain
| | - Dominique Charron
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-97f, Université Paris-Diderot, HLA et Médecine, Labex Transplantex, Hôpital Saint-Louis, Paris, France
| | - Francisco Fernández-Avilés
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Madrid, Spain
| | | |
Collapse
|
32
|
Fernández-García V, González-Ramos S, Martín-Sanz P, García-Del Portillo F, Laparra JM, Boscá L. NOD1 in the interplay between microbiota and gastrointestinal immune adaptations. Pharmacol Res 2021; 171:105775. [PMID: 34273489 DOI: 10.1016/j.phrs.2021.105775] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1), a pattern recognition receptor (PRR) that detects bacterial peptidoglycan fragments and other danger signals, has been linked to inflammatory pathologies. NOD1, which is expressed by immune and non-immune cells, is activated after recognizing microbe-associated molecular patterns (MAMPs). This recognition triggers host defense responses and both immune memory and tolerance can also be achieved during these processes. Since the gut microbiota is currently considered a master regulator of human physiology central in health and disease and the intestine metabolizes a wide range of nutrients, drugs and hormones, it is a fact that dysbiosis can alter tissues and organs homeostasis. These systemic alterations occur in response to gastrointestinal immune adaptations that are not yet fully understood. Even if previous evidence confirms the connection between the microbiota, the immune system and metabolic disorders, much remains to be discovered about the contribution of NOD1 to low-grade inflammatory pathologies such as obesity, diabetes and cardiovascular diseases. This review compiles the most recent findings in this area, while providing a dynamic and practical framework with future approaches for research and clinical applications on targeting NOD1. This knowledge can help to rate the consequences of the disease and to stratify the patients for therapeutic interventions.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | | | - José Moisés Laparra
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra, Cantoblanco 8, 28049 Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
33
|
Fernández-García V, González-Ramos S, Martín-Sanz P, Laparra JM, Boscá L. Beyond classic concepts in thyroid homeostasis: Immune system and microbiota. Mol Cell Endocrinol 2021; 533:111333. [PMID: 34048865 DOI: 10.1016/j.mce.2021.111333] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
It has long been known that thyroid hormones have implications for multiple physiological processes and can lead to serious illness when there is an imbalance in its metabolism. The connections between thyroid hormone metabolism and the immune system have been extensively described, as they can participate in inflammation, autoimmunity, or cancer progression. In addition, changes in the normal intestinal microbiota involve the activation of the immune system while triggering different pathophysiological disorders. Recent studies have linked the microbiota and certain bacterial fragments or metabolites to the regulation of thyroid hormones and the general response in the endocrine system. Even if the biology and function of the thyroid gland has attracted more attention due to its pathophysiological importance, there are essential mechanisms and issues related to it that are related to the interplay between the intestinal microbiota and the immune system and must be further investigated. Here we summarize additional information to uncover these relationships, the knowledge of which would help establish new personalized medical strategies.
Collapse
Affiliation(s)
- Victoria Fernández-García
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain
| | - Silvia González-Ramos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain.
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Melchor Fernández Almagro 6, 28029, Madrid, Spain
| | - José M Laparra
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra. Cantoblanco 8, 28049, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029, Madrid, Spain.
| |
Collapse
|
34
|
Ahmadi A, Vahabzadeh Z, Moloudi M, Farhadi L, Shirahmadi S. Contribution of toll-like receptor 2 and nicotinamide adenine dinucleotide phosphate oxidase to the trimethylamine N-oxide-induced inflammatory reactions in U937-derived macrophages. ARYA ATHEROSCLEROSIS 2021; 17:1-7. [PMID: 35685229 PMCID: PMC9137230 DOI: 10.22122/arya.v17i0.2096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/24/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is emerging as a new generation of metabolites related to the activation of inflammatory reactions in the macrophages during atherosclerosis. Stress-activation of cell surface toll-like receptors (TLRs) as well as nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) is also assumed to be involved in TMAO-induced inflammatory reaction in the macrophages. To elucidate the possible contribution of TLRs and NOX to the mentioned signaling pathway, we aimed to simultaneously evaluate the expression level of TLR2, TLR6, and NOX2 in TMAO-treated macrophages. METHODS 2.5 × 106 cells of U937-derived macrophages were treated in triplicates with different concentrations (37.5, 75, 150, and 300 μM) of TMAO for 24 hours. The cells were also treated with tunicamycin (TUN), as a positive control of stress. Normal control group (CTR) cells received no treatment. The viability of treated cells was checked by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, a tetrazole (MTT) assay. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was also used to evaluate the relative expression (fold change) of TLR2, TLR6, and NOX2 at messenger ribonucleic acid (mRNA) levels. One-way analysis of variance (ANOVA) with post-hoc Dunnett's test was performed to compare every mean with that of the control. RESULTS No cell death occurred because of treatments. Dose of 300 μM of TMAO significantly increased the relative expression of both TLR2 and NOX2 compared to the CTR cells (P < 0.001 for both). The elevation of TLR6 was not statistically significant in all groups of TMAO-treated cells (P > 0.050). CONCLUSION Our results provide documentation supporting contribution of TLR2 and NOX2 to previously described inflammatory reactions induced by TMAO in macrophages. In addition, they may clarify the proatherogenic role of TMAO in foam cell formation as well as abnormal activation of macrophages during atherosclerosis.
Collapse
Affiliation(s)
- Abbas Ahmadi
- Assistant Professor, Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Zakaria Vahabzadeh
- Assistant Professor, Liver and Digestive Research Center, Research Institute for Health Development AND Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammadraman Moloudi
- Assistant Professor, Liver and Digestive Research Center, Research Institute for Health Development AND Department of Medical Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Leila Farhadi
- PhD Candidate, Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sara Shirahmadi
- Medical Student, Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
35
|
Li Q, Gao B, Siqin B, He Q, Zhang R, Meng X, Zhang N, Zhang N, Li M. Gut Microbiota: A Novel Regulator of Cardiovascular Disease and Key Factor in the Therapeutic Effects of Flavonoids. Front Pharmacol 2021; 12:651926. [PMID: 34220497 PMCID: PMC8241904 DOI: 10.3389/fphar.2021.651926] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/23/2021] [Indexed: 01/30/2023] Open
Abstract
Cardiovascular disease is the main cause of death worldwide, and traditional cardiovascular risk factors cannot fully explain the occurrence of the disease. In recent years, the relationship between gut microbiota and its metabolites and cardiovascular disease has been a hot study topic. The changes in gut microbiota and its metabolites are related to the occurrence and development of atherosclerosis, myocardial infarction, heart failure, and hypertension. The mechanisms by which gut microbiota and its metabolites influence cardiovascular disease have been reported, although not comprehensively. Additionally, following ingestion, flavonoids are decomposed into phenolic acids that are more easily absorbed by the body after being processed by enzymes produced by intestinal microorganisms, which increases flavonoid bioavailability and activity, consequently affecting the onset of cardiovascular disease. However, flavonoids can also inhibit the growth of harmful microorganisms, promote the proliferation of beneficial microorganisms, and maintain the balance of gut microbiota. Hence, it is important to study the relationship between gut microbiota and flavonoids to elucidate the protective effects of flavonoids in cardiovascular diseases. This article will review the role and mechanism of gut microbiota and its metabolites in the occurrence and development of atherosclerosis, myocardial infarction, heart failure, and hypertension. It also discusses the potential value of flavonoids in the prevention and treatment of cardiovascular disease following their transformation through gut microbiota metabolism.
Collapse
Affiliation(s)
- Qinyu Li
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Bing Gao
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Bateer Siqin
- Xilinguole Meng Mongolian General Hospital, Xilinhaote, China
| | - Qian He
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Ru Zhang
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Xiangxi Meng
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Naiheng Zhang
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Na Zhang
- Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Minhui Li
- Department of Pharmacy, Baotou Medical College, Baotou, China
- Pharmaceutical Laboratory, Inner Mongolia Institute of Traditional Chinese Medicine, Hohhot, China
- Inner Mongolia Key Laboratory of Characteristic Geoherbs Resources and Utilization, Baotou Medical College, Baotou, China
- Office of Academic Research, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
36
|
Probiotics supplementation and cardiometabolic risk factors: A new insight into recent advances, potential mechanisms, and clinical implications. PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
37
|
Garshick MS, Nikain C, Tawil M, Pena S, Barrett TJ, Wu BG, Gao Z, Blaser MJ, Fisher EA. Reshaping of the gastrointestinal microbiome alters atherosclerotic plaque inflammation resolution in mice. Sci Rep 2021; 11:8966. [PMID: 33903700 PMCID: PMC8076321 DOI: 10.1038/s41598-021-88479-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Since alterations in the intestinal microbiota may induce systemic inflammation and polarization of macrophages to the M1 state, the microbiome role in atherosclerosis, an M1-driven disease, requires evaluation. We aimed to determine if antibiotic (Abx) induced alterations to the intestinal microbiota interferes with atherosclerotic plaque inflammation resolution after lipid-lowering in mice. Hyperlipidemic Apoe−/− mice were fed a western diet to develop aortic atherosclerosis with aortas then transplanted into normolipidemic wild-type (WT) mice to model clinically aggressive lipid management and promote atherosclerosis inflammation resolution. Gut microbial composition pre and post-transplant was altered via an enteral antibiotic or not. Post aortic transplant, after Abx treatment, while plaque size did not differ, compared to Apoe−/− mice, Abx– WT recipient mice had a 32% reduction in CD68-expressing cells (p = 0.02) vs. a non-significant 12% reduction in Abx+ WT mice. A trend toward an M1 plaque CD68-expresing cell phenotype was noted in Abx+ mice. By 16S rRNA sequence analysis, the Abx+ mice had reduced alpha diversity and increased Firmicutes/Bacteroidetes relative abundance ratio with a correlation between gut Firmicutes abundance and plaque CD68-expressing cell content (p < 0.05). These results indicate that in a murine atherosclerotic plaque inflammation resolution model, antibiotic-induced microbiome perturbation may blunt the effectiveness of lipid-lowering to reduce the content of plaque inflammatory CD68-expressing cells.
Collapse
Affiliation(s)
- Michael S Garshick
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine, New York, USA.,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Cyrus Nikain
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Michael Tawil
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Stephanie Pena
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Tessa J Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Benjamin G Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University School of Medicine, New York, USA.,Division of Pulmonary and Critical Care, Veterans Affairs New York Harbor Healthcare System, New York, NY, USA
| | - Zhan Gao
- Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane West, Piscataway, NJ, 08854, USA.
| | - Edward A Fisher
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine, New York, USA. .,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA. .,Marc and Ruti Bell Vascular Biology Program, Cardiovascular Research Center, New York University Langone Health, New York, USA.
| |
Collapse
|
38
|
Xue J, Allaband C, Zhou D, Poulsen O, Martino C, Jiang L, Tripathi A, Elijah E, Dorrestein PC, Knight R, Zarrinpar A, Haddad GG. Influence of Intermittent Hypoxia/Hypercapnia on Atherosclerosis, Gut Microbiome, and Metabolome. Front Physiol 2021; 12:663950. [PMID: 33897472 PMCID: PMC8060652 DOI: 10.3389/fphys.2021.663950] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/17/2021] [Indexed: 01/05/2023] Open
Abstract
Obstructive sleep apnea (OSA), a common sleep disorder characterized by intermittent hypoxia and hypercapnia (IHC), increases atherosclerosis risk. However, the contribution of intermittent hypoxia (IH) or intermittent hypercapnia (IC) in promoting atherosclerosis remains unclear. Since gut microbiota and metabolites have been implicated in atherosclerosis, we examined whether IH or IC alters the microbiome and metabolome to induce a pro-atherosclerotic state. Apolipoprotein E deficient mice (ApoE-/- ), treated with IH or IC on a high-fat diet (HFD) for 10 weeks, were compared to Air controls. Atherosclerotic lesions were examined, gut microbiome was profiled using 16S rRNA gene amplicon sequencing and metabolome was assessed by untargeted mass spectrometry. In the aorta, IC-induced atherosclerosis was significantly greater than IH and Air controls (aorta, IC 11.1 ± 0.7% vs. IH 7.6 ± 0.4%, p < 0.05 vs. Air 8.1 ± 0.8%, p < 0.05). In the pulmonary artery (PA), however, IH, IC, and Air were significantly different from each other in atherosclerotic formation with the largest lesion observed under IH (PA, IH 40.9 ± 2.0% vs. IC 20.1 ± 2.6% vs. Air 12.2 ± 1.5%, p < 0.05). The most differentially abundant microbial families (p < 0.001) were Peptostreptococcaceae, Ruminococcaceae, and Erysipelotrichaceae. The most differentially abundant metabolites (p < 0.001) were tauro-β-muricholic acid, ursodeoxycholic acid, and lysophosphoethanolamine (18:0). We conclude that IH and IC (a) modulate atherosclerosis progression differently in distinct vascular beds with IC, unlike IH, facilitating atherosclerosis in both aorta and PA and (b) promote an atherosclerotic luminal gut environment that is more evident in IH than IC. We speculate that the resulting changes in the gut metabolome and microbiome interact differently with distinct vascular beds.
Collapse
Affiliation(s)
- Jin Xue
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - Celeste Allaband
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Biomedical Sciences Program, University of California, San Diego, San Diego, CA, United States
- Division of Gastroenterology, University of California, San Diego, San Diego, CA, United States
| | - Dan Zhou
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - Orit Poulsen
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - Cameron Martino
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Bioinformatics and Systems Biology Program, University of California, San Diego, San Diego, CA, United States
- Center for Microbiome Innovation, University of California, San Diego, San Diego, CA, United States
| | - Lingjing Jiang
- Division of Biostatistics, University of California, San Diego, San Diego, CA, United States
| | - Anupriya Tripathi
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, CA, United States
| | - Emmanuel Elijah
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, CA, United States
- Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, San Diego, CA, United States
| | - Pieter C. Dorrestein
- Center for Microbiome Innovation, University of California, San Diego, San Diego, CA, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, CA, United States
- Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, San Diego, CA, United States
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Center for Microbiome Innovation, University of California, San Diego, San Diego, CA, United States
- Department of Computer Science and Engineering, University of California, San Diego, San Diego, CA, United States
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, San Diego, CA, United States
- Center for Microbiome Innovation, University of California, San Diego, San Diego, CA, United States
- Division of Gastroenterology, VA San Diego, La Jolla, CA, United States
- Institute of Diabetes and Metabolic Health, University of California, San Diego, San Diego, CA, United States
| | - Gabriel G. Haddad
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Department of Neuroscience, University of California, San Diego, San Diego, CA, United States
- Rady Children’s Hospital, San Diego, CA, United States
| |
Collapse
|
39
|
Mangalam AK, Yadav M, Yadav R. The Emerging World of Microbiome in Autoimmune Disorders: Opportunities and Challenges. INDIAN JOURNAL OF RHEUMATOLOGY 2021; 16:57-72. [PMID: 34531642 PMCID: PMC8442979 DOI: 10.4103/injr.injr_210_20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Trillions of commensal bacteria colonizing humans (microbiome) have emerged as essential player(s) in human health. The alteration of the same has been linked with diseases including autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, and ankylosing spondylitis. Gut bacteria are separated from the host through a physical barrier such as skin or gut epithelial lining. However, the perturbation in the healthy bacterial community (gut dysbiosis) can compromise gut barrier integrity, resulting in translocation of bacterial contents across the epithelial barrier (leaky gut). Bacterial contents such as lipopolysaccharide and bacterial antigens can induce a systemic inflammatory environment through activation and induction of immune cells. The biggest question in the field is whether inflammation causes gut dysbiosis or dysbiosis leads to disease induction or propagation, i.e., it is inside out or outside in or both. In this review, we first discuss the microbiome profiling studies in various autoimmune disorders, followed by a discussion of potential mechanisms through which microbiome is involved in the pathobiology of diseases. A better understanding of the role of the microbiome in health and disease will help us harness the power of commensal bacteria for the development of novel therapeutic agents to treat autoimmune disorders.
Collapse
Affiliation(s)
| | - Meeta Yadav
- Department of Pathology, University of Iowa, Iowa, IA,
USA
| | - Rajwardhan Yadav
- Department of Rheumatology, St Francis Hospital, Hartford,
CT, USA
| |
Collapse
|
40
|
Nabrdalik K, Krzyżak K, Hajzler W, Drożdż K, Kwiendacz H, Gumprecht J, Lip GYH. Fat, Sugar or Gut Microbiota in Reducing Cardiometabolic Risk: Does Diet Type Really Matter? Nutrients 2021; 13:639. [PMID: 33669342 PMCID: PMC7920316 DOI: 10.3390/nu13020639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence of cardiometabolic diseases, such as obesity, diabetes, and cardiovascular diseases, is constantly rising. Successful lifestyle changes may limit their incidence, which is why researchers focus on the role of nutrition in this context. The outcomes of studies carried out in past decades have influenced dietary guidelines, which primarily recommend reducing saturated fat as a therapeutic approach for cardiovascular disease prevention, while limiting the role of sugar due to its harmful effects. On the other hand, a low-carbohydrate diet (LCD) as a method of treatment remains controversial. A number of studies on the effect of LCDs on patients with type 2 diabetes mellitus proved that it is a safe and effective method of dietary management. As for the risk of cardiovascular diseases, the source of carbohydrates and fats corresponds with the mortality rate and protective effect of plant-derived components. Additionally, some recent studies have focused on the gut microbiota in relation to cardiometabolic diseases and diet as one of the leading factors affecting microbiota composition. Unfortunately, there is still no precise answer to the question of which a single nutrient plays the most important role in reducing cardiometabolic risk, and this review article presents the current state of the knowledge in this field.
Collapse
Affiliation(s)
- Katarzyna Nabrdalik
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool L14 3PE, UK;
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.D.); (H.K.); (J.G.)
| | - Katarzyna Krzyżak
- Students’ Scientific Association by the Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.K.); (W.H.)
| | - Weronika Hajzler
- Students’ Scientific Association by the Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.K.); (W.H.)
| | - Karolina Drożdż
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.D.); (H.K.); (J.G.)
| | - Hanna Kwiendacz
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.D.); (H.K.); (J.G.)
| | - Janusz Gumprecht
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.D.); (H.K.); (J.G.)
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool L14 3PE, UK;
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (K.D.); (H.K.); (J.G.)
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, 9100 Aalborg, Denmark
| |
Collapse
|
41
|
Samadi A, Sabuncuoglu S, Samadi M, Isikhan SY, Chirumbolo S, Peana M, Lay I, Yalcinkaya A, Bjørklund G. A Comprehensive Review on Oxysterols and Related Diseases. Curr Med Chem 2021; 28:110-136. [PMID: 32175830 DOI: 10.2174/0929867327666200316142659] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/09/2019] [Accepted: 11/10/2019] [Indexed: 11/22/2022]
Abstract
The present review aims to provide a complete and comprehensive summary of current literature relevant to oxysterols and related diseases. Oxidation of cholesterol leads to the formation of a large number of oxidized products, generally known as oxysterols. They are intermediates in the biosynthesis of bile acids, steroid hormones, and 1,25- dihydroxyvitamin D3. Although oxysterols are considered as metabolic intermediates, there is a growing body of evidence that many of them are bioactive, and their absence or excess may be part of the cause of a disease phenotype. These compounds derive from either enzymatic or non-enzymatic oxidation of cholesterol. This study provides comprehensive information about the structures, formation, and types of oxysterols even when involved in certain disease states, focusing on their effects on metabolism and linkages with these diseases. The role of specific oxysterols as mediators in various disorders, such as degenerative (age-related) and cancer-related disorders, has now become clearer. Oxysterol levels may be employed as suitable markers for the diagnosis of specific diseases or in predicting the incidence rate of diseases, such as diabetes mellitus, Alzheimer's disease, multiple sclerosis, osteoporosis, lung cancer, breast cancer, and infertility. However, further investigations may be required to confirm these mentioned possibilities.
Collapse
Affiliation(s)
- Afshin Samadi
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Suna Sabuncuoglu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Mahshid Samadi
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Selen Yilmaz Isikhan
- Vocational Higher School of Social Sciences, Hacettepe University, Ankara, Turkey
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Incilay Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ahmet Yalcinkaya
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
42
|
Huang L, Wang J, Xu R, Liu Y, Liu Z. Regulatory effect of traditional Chinese medicine on gut microbiota in patients with atherosclerosis: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e23730. [PMID: 33327364 PMCID: PMC7738117 DOI: 10.1097/md.0000000000023730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Atherosclerosis is the pathological basis of many cardiovascular and cerebrovascular diseases, and its pathogenesis is complex. Recent studies revealed a significant role of gut microbiota in the onset and development of atherosclerosis. Traditional Chinese medicine has rich clinical experience and unique advantages in the treatment of atherosclerosis. A large number of studies have proved that traditional Chinese medicine has the functions of reducing blood lipid, regulating gut microbiota, and resisting inflammation. The aim of this systematic review is to observe the randomized controlled trial of traditional Chinese medicine in treating gut microbiota, so as to evaluate the effectiveness and safety of traditional Chinese medicine in treating atherosclerosis patients. METHODS The English database (PubMed, Web of Science, Embase, the Cochrane Library) and Chinese database (China National Knowledge Infrastructure, the Chongqing VIP Chinese Science, and Technology Periodic Database, Wanfang Database, and China Biomedical Literature Database) will be searched up to October 2020. We will also manually search the Chinese clinical trial register, conference papers, and unpublished studies or references. Randomized control trials of traditional Chinese medicine treatment of atherosclerosis were collected comprehensively, and 2 researchers will independently screen literature, data extraction, and evaluation the quality of literature methodology. The primary outcomes are lipid metabolism and gut microbiota and their metabolites. The secondary outcomes are the change of inflammatory markers. Meta-analysis was performed by RevMan 5.3.5 software. The Grades of Recommendation, Assessment, Development, and Evaluation will be used to evaluate the outcome quality of evidence. RESULTS This study will comprehensively review the existing evidence of traditional Chinese medicine in treating atherosclerosis from the perspective of gut microbiota. CONCLUSION This study will provide information on the effectiveness and safety of traditional Chinese medicine in treating atherosclerosis from the perspective of gut microbiota. UNIQUE INPLASY NUMBER INPLASY2020110056.
Collapse
Affiliation(s)
| | - Jianan Wang
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, PR China
| | - Ri Xu
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, PR China
| | - Yanwei Liu
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, PR China
| | - Zhongyong Liu
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, PR China
| |
Collapse
|
43
|
Mutual Interplay of Host Immune System and Gut Microbiota in the Immunopathology of Atherosclerosis. Int J Mol Sci 2020; 21:ijms21228729. [PMID: 33227973 PMCID: PMC7699263 DOI: 10.3390/ijms21228729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is the key for the initiation and progression of atherosclerosis. Accumulating evidence has revealed that an altered gut microbiome (dysbiosis) triggers both local and systemic inflammation to cause chronic inflammatory diseases, including atherosclerosis. There have been some microbiome-relevant pro-inflammatory mechanisms proposed to link the relationships between dysbiosis and atherosclerosis such as gut permeability disruption, trigger of innate immunity from lipopolysaccharide (LPS), and generation of proatherogenic metabolites, such as trimethylamine N-oxide (TMAO). Meanwhile, immune responses, such as inflammasome activation and cytokine production, could reshape both composition and function of the microbiota. In fact, the immune system delicately modulates the interplay between microbiota and atherogenesis. Recent clinical trials have suggested the potential of immunomodulation as a treatment strategy of atherosclerosis. Here in this review, we present current knowledge regarding to the roles of microbiota in contributing atherosclerotic pathogenesis and highlight translational perspectives by discussing the mutual interplay between microbiota and immune system on atherogenesis.
Collapse
|
44
|
Abstract
Innate lymphoid cells (ILCs) are a large family of cells of the immune system that performs various functions in immune defense, inflammation, and tissue remodeling. As a part of the innate immune system, ILCs are a distinct form of lymphocytes different from T and B cells. ILCs can provide host defense against the source of infection and initiate the repair and remodeling processes to restore and maintain host body homeostasis. The number of patients with Crohn’s disease (CD) worldwide has continued to increase in recent years and this disease has brought sickness and death to many families. Numerous studies have found that ILCs also undergo a series of alternations during the development of CD and contribute to this disease. Despite this, the pathogenesis of CD is still not fully explained. So, we keep researching and exploring. In this review, we have closely linked the latest progress on ILCs and CD, and introduced, in detail, the specific roles of four different types of ILCs in CD. We also describe new progress in the pathogenesis of CD, with particular emphasis on the plasticity of ILC3s in this disease. These new studies and findings may provide new insights and breakthrough points for the treatment of CD.
Collapse
Affiliation(s)
- Ying Wu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
45
|
Cavadas B, Camacho R, Ferreira JC, Ferreira RM, Figueiredo C, Brazma A, Fonseca NA, Pereira L. Gastric Microbiome Diversities in Gastric Cancer Patients from Europe and Asia Mimic the Human Population Structure and Are Partly Driven by Microbiome Quantitative Trait Loci. Microorganisms 2020; 8:microorganisms8081196. [PMID: 32781641 PMCID: PMC7463948 DOI: 10.3390/microorganisms8081196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/15/2022] Open
Abstract
The human gastrointestinal tract harbors approximately 100 trillion microorganisms with different microbial compositions across geographic locations. In this work, we used RNASeq data from stomach samples of non-disease (164 individuals from European ancestry) and gastric cancer patients (137 from Europe and Asia) from public databases. Although these data were intended to characterize the human expression profiles, they allowed for a reliable inference of the microbiome composition, as confirmed from measures such as the genus coverage, richness and evenness. The microbiome diversity (weighted UniFrac distances) in gastric cancer mimics host diversity across the world, with European gastric microbiome profiles clustering together, distinct from Asian ones. Despite the confirmed loss of microbiome diversity from a healthy status to a cancer status, the structured profile was still recognized in the disease condition. In concordance with the parallel host-bacteria population structure, we found 16 human loci (non-synonymous variants) in the European-descendent cohorts that were significantly associated with specific genera abundance. These microbiome quantitative trait loci display heterogeneity between population groups, being mainly linked to the immune system or cellular features that may play a role in enabling microbe colonization and inflammation.
Collapse
Affiliation(s)
- Bruno Cavadas
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.C.F.); (R.M.F.); (C.F.); (L.P.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Correspondence:
| | - Rui Camacho
- FEUP-Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal;
- INESC TEC—Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, Universidade do Porto, 4200-465 Porto, Portugal
| | - Joana C. Ferreira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.C.F.); (R.M.F.); (C.F.); (L.P.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rui M. Ferreira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.C.F.); (R.M.F.); (C.F.); (L.P.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ceu Figueiredo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.C.F.); (R.M.F.); (C.F.); (L.P.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, 4200-135 Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Alvis Brazma
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK;
| | - Nuno A. Fonseca
- CIBIO—Centro de Investigação em Biodiversidade e Recursos Genético, Universidade do Porto, 4485-661 Vairão, Portugal;
| | - Luísa Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (J.C.F.); (R.M.F.); (C.F.); (L.P.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, 4200-135 Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| |
Collapse
|
46
|
Abstract
We critically review potential involvement of trimethylamine N-oxide (TMAO) as a link between diet, the gut microbiota and CVD. Generated primarily from dietary choline and carnitine by gut bacteria and hepatic flavin-containing mono-oxygenase (FMO) activity, TMAO could promote cardiometabolic disease when chronically elevated. However, control of circulating TMAO is poorly understood, and diet, age, body mass, sex hormones, renal clearance, FMO3 expression and genetic background may explain as little as 25 % of TMAO variance. The basis of elevations with obesity, diabetes, atherosclerosis or CHD is similarly ill-defined, although gut microbiota profiles/remodelling appear critical. Elevated TMAO could promote CVD via inflammation, oxidative stress, scavenger receptor up-regulation, reverse cholesterol transport (RCT) inhibition, and cardiovascular dysfunction. However, concentrations influencing inflammation, scavenger receptors and RCT (≥100 µm) are only achieved in advanced heart failure or chronic kidney disease (CKD), and greatly exceed pathogenicity of <1-5 µm levels implied in some TMAO-CVD associations. There is also evidence that CVD risk is insensitive to TMAO variance beyond these levels in omnivores and vegetarians, and that major TMAO sources are cardioprotective. Assessing available evidence suggests that modest elevations in TMAO (≤10 µm) are a non-pathogenic consequence of diverse risk factors (ageing, obesity, dyslipidaemia, insulin resistance/diabetes, renal dysfunction), indirectly reflecting CVD risk without participating mechanistically. Nonetheless, TMAO may surpass a pathogenic threshold as a consequence of CVD/CKD, secondarily promoting disease progression. TMAO might thus reflect early CVD risk while providing a prognostic biomarker or secondary target in established disease, although mechanistic contributions to CVD await confirmation.
Collapse
|
47
|
Massmig M, Reijerse E, Krausze J, Laurich C, Lubitz W, Jahn D, Moser J. Carnitine metabolism in the human gut: characterization of the two-component carnitine monooxygenase CntAB from Acinetobacter baumannii. J Biol Chem 2020; 295:13065-13078. [PMID: 32694223 DOI: 10.1074/jbc.ra120.014266] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/24/2020] [Indexed: 01/29/2023] Open
Abstract
Bacterial formation of trimethylamine (TMA) from carnitine in the gut microbiome has been linked to cardiovascular disease. During this process, the two-component carnitine monooxygenase (CntAB) catalyzes the oxygen-dependent cleavage of carnitine to TMA and malic semialdehyde. Individual redox states of the reductase CntB and the catalytic component CntA were investigated based on mutagenesis and electron paramagnetic resonance (EPR) spectroscopic approaches. Protein ligands of the flavin mononucleotide (FMN) and the plant-type [2Fe-2S] cluster of CntB and also of the Rieske-type [2Fe-2S] cluster and the mononuclear [Fe] center of CntA were identified. EPR spectroscopy of variant CntA proteins suggested a hierarchical metallocenter maturation, Rieske [2Fe-2S] followed by the mononuclear [Fe] center. NADH-dependent electron transfer via the redox components of CntB and within the trimeric CntA complex for the activation of molecular oxygen was investigated. EPR experiments indicated that the two electrons from NADH were allocated to the plant-type [2Fe-2S] cluster and to FMN in the form of a flavin semiquinone radical. Single-turnover experiments of this reduced CntB species indicated the translocation of the first electron onto the [Fe] center and the second electron onto the Rieske-type [2Fe-2S] cluster of CntA to finally allow for oxygen activation as a basis for carnitine cleavage. EPR spectroscopic investigation of CntA variants indicated an unusual intermolecular electron transfer between the subunits of the CntA trimer via the "bridging" residue Glu-205. On the basis of these data, a redox catalytic cycle for carnitine monooxygenase was proposed.
Collapse
Affiliation(s)
- Marco Massmig
- Institute of Microbiology, Technical University Braunschweig, Braunschweig, Germany
| | - Edward Reijerse
- Max-Planck-Institute for Chemical Energy Conversion, Mülheim an der Ruhr, Germany
| | - Joern Krausze
- Institute of Plant Biology, Technical University Braunschweig, Braunschweig, Germany
| | - Christoph Laurich
- Max-Planck-Institute for Chemical Energy Conversion, Mülheim an der Ruhr, Germany
| | - Wolfgang Lubitz
- Max-Planck-Institute for Chemical Energy Conversion, Mülheim an der Ruhr, Germany
| | - Dieter Jahn
- Braunschweig Centre of Integrated Systems Biology, Braunschweig, Germany
| | - Jürgen Moser
- Institute of Microbiology, Technical University Braunschweig, Braunschweig, Germany.
| |
Collapse
|
48
|
Torchia MT, Amakiri I, Werth P, Moschetti W. Characterization of native knee microorganisms using next-generation sequencing in patients undergoing primary total knee arthroplasty. Knee 2020; 27:1113-1119. [PMID: 31926670 DOI: 10.1016/j.knee.2019.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/18/2019] [Accepted: 12/17/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Next-generation sequencing (NGS) offers improved sensitivity compared to culture-based methods for identifying organisms from synovial joints. It remains unclear whether native microorganisms exist in a joint, and positive NGS results may be interpreted as pathologic when in fact they may represent this native microbiome. The purpose of this study was to characterize the native knee microorganism profile in patients undergoing primary total knee arthroplasty (TKA). METHODS Forty consecutive patients with osteoarthritis undergoing primary total knee arthroplasty were enrolled prospectively. During TKA surgery but prior to arthrotomy, the native knee was aspirated and the fluid was sent for NGS analysis. Immediately after arthrotomy, four separate tissue samples were also sent for NGS analysis. All microbes identified by NGS were recorded. RESULTS Twelve out of forty patients (30%) had at least one positive organism identified by NGS from their native knee. Of those with positive NGS results, 9/12 (75%) had more than one organism identified (range two to 11). There were no significant differences in demographics, comorbidities, or incidence of prior knee injections between the two groups. There were 48 unique organisms identified from all patients, and the average number of organisms identified by NGS was 4.6 per patient. Four sterile water controls were all negative for organisms. CONCLUSION A proportion of patients with osteoarthritis undergoing primary total knee arthroplasty have organisms identified in their joint by NGS at the time of surgery. Organisms identified after TKA by NGS when concern for periprosthetic joint infection exists may represent the native microbiome rather than pathogenic microbes.
Collapse
Affiliation(s)
- Michael T Torchia
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA.
| | - Ikechukwu Amakiri
- Department of Orthopaedics, Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| | - Paul Werth
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Wayne Moschetti
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA; Department of Orthopaedics, Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH 03755, USA
| |
Collapse
|
49
|
Lieshchova MA, Bilan MV, Bohomaz AA, Tishkina NM, Brygadyrenko VV. Effect of succinic acid on the organism of mice and their intestinal microbiota against the background of excessive fat consumption. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Succinic acid and its salts (succinates) positively affect the oxygenation of the internal environment, stabilize the structure and functional activity of mitochondria, and normalize the ion metabolism in the cell. Separate clinical studies and experimental surveys confirmed that having low toxicity succinic acid has well-manifested antioxidant, immunostimulating, adaptogenic properties. In this study, we determined the influence of succinic acid on the organism of laboratory animals against the backround of high-fat diet: the changes in body weight, indices of the mass of the internal organs, blood parameters and the changes in the intestinal microbiota were determined. For the experiment, we formed three experimental and three control groups of male white mice. The animals of the control group received 0.5% solution of succinic acid instead of water. In the experiment, we determined that succinic acid has no effect on the intensity of growth of weight of young mice against the background of excessive fat in their diet. Excessive consumption of fat by male mice leads to mainly disorders in the functioning of the liver, excretory and the immune systems. High-fat diet of mice is accompanied by impaired hepatic function, manifested in sharp hypoproteinemia due to globulins, increase in the activity of hepatic enzymes against the background of reduced activity of alkaline phosphatase, increase in the level of bilirubin, and decrease in glucose. Excess of fat in the diet leads to malfunctioning of the excretory system, manifested in the reduced index of kidneys’ weight, high content of creatinine and reduced level of urea in the blood. Addition of succinic acid has a positive effect on the functional condition of the liver and the kidneys, especially noticeable during long-term intake. High-fat diet causes disorders in the functioning of the organs of blood circulation and immune protection, accompanied by decrease in the relative mass of the thymus and spleen, low content of hemoglobin and the number of erythrocytes, but has no significant effect on the content of other cellular elements in the blood. By the middle of the experiment, succinic acid had exacerbated these processes compared to the control, but by the end of the experiment, by contrast, these processes were alleviated. Addition of the succinic acid to high-fat diet contributed to the change in the quantitative composition of the main representatives of the obligatory microbiota (Bifidobacterium spp., Lactobacillus spp. and typical Escherichia coli) in the laboratory animals. Such changes in the intestinal microbiota may lead to such consequences as reproduction of the facultative microflora, and, thus, development of various diseases.
Collapse
|
50
|
Kazemian N, Mahmoudi M, Halperin F, Wu JC, Pakpour S. Gut microbiota and cardiovascular disease: opportunities and challenges. MICROBIOME 2020; 8:36. [PMID: 32169105 PMCID: PMC7071638 DOI: 10.1186/s40168-020-00821-0] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 03/02/2020] [Indexed: 05/03/2023]
Abstract
Coronary artery disease (CAD) is the most common health problem worldwide and remains the leading cause of morbidity and mortality. Over the past decade, it has become clear that the inhabitants of our gut, the gut microbiota, play a vital role in human metabolism, immunity, and reactions to diseases, including CAD. Although correlations have been shown between CAD and the gut microbiota, demonstration of potential causal relationships is much more complex and challenging. In this review, we will discuss the potential direct and indirect causal roots between gut microbiota and CAD development via microbial metabolites and interaction with the immune system. Uncovering the causal relationship of gut microbiota and CAD development can lead to novel microbiome-based preventative and therapeutic interventions. However, an interdisciplinary approach is required to shed light on gut bacterial-mediated mechanisms (e.g., using advanced nanomedicine technologies and incorporation of demographic factors such as age, sex, and ethnicity) to enable efficacious and high-precision preventative and therapeutic strategies for CAD.
Collapse
Affiliation(s)
- Negin Kazemian
- School of Engineering, University of British Columbia, Kelowna, Kelowna, BC, Canada
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA.
| | | | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sepideh Pakpour
- School of Engineering, University of British Columbia, Kelowna, Kelowna, BC, Canada.
| |
Collapse
|