1
|
Petersen C, Satheesh Babu AK, Della Lucia CM, Paz HA, Iglesias-Carres L, Zhong Y, Jalili T, Symons JD, Shankar K, Neilson AP, Wankhade UD, Anandh Babu PV. Gut microbes metabolize strawberry phytochemicals and mediate their beneficial effects on vascular inflammation. Gut Microbes 2025; 17:2446375. [PMID: 39760464 DOI: 10.1080/19490976.2024.2446375] [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: 03/20/2024] [Revised: 11/02/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Evidence suggests that a healthy gut microbiome is essential for metabolizing dietary phytochemicals. However, the microbiome's role in metabolite production and the influence of gut dysbiosis on this process remain unclear. Further, studies on the relationship among gut microbes, metabolites, and biological activities of phytochemicals are limited. We addressed this knowledge gap using strawberry phytochemicals as a model. C57BL/6J mice were fed a standard diet [C]; strawberry-supplemented diet (~2 human servings) [CS]; strawberry-supplemented diet and treated with antibiotics (to deplete gut microbes) [CSA]; high-fat diet (HFD) [HF]; strawberry-supplemented HFD [HS]; and strawberry-supplemented HFD and treated with antibiotics [HSA] for 12 weeks. First, antibiotic treatment suppressed the production of selected metabolites (CSA vs. CS), and p-coumaric acid was identified as a strawberry-derived microbial metabolite. Second, HFD-induced dysbiosis negatively affected metabolite production (HS vs. HF), and hippuric acid was identified as a microbial metabolite in HFD conditions. Third, dietary strawberries improved HFD-induced vascular inflammation (HS vs. HF). However, antibiotic treatment reduced metabolite production and abolished the vascular effects of strawberries (HSA vs. HS), indicating the importance of gut microbes in mediating the vascular benefits of strawberries via metabolites. Fourth, strawberry supplementation decreased Coprobacillus that was positively associated with vascular inflammation, whereas it increased Lachnospiraceae that was negatively associated with vascular inflammation and positively associated with hippuric acid. Fifth, hippuric acid was negatively associated with vascular inflammation. Our study fills in some pieces of the giant puzzle regarding the influence of gut microbes on the biological activities of phytochemicals. HFD-induced gut dysbiosis negatively impacts metabolite production and a strong association exists among gut microbes, strawberry-derived microbial metabolites, and the vascular benefits of dietary strawberries. Further, our study provides significant proof of concept to warrant future research on the use of strawberries as a nutritional strategy to prevent vascular complications.
Collapse
Affiliation(s)
- Chrissa Petersen
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | | | - Ceres Mattos Della Lucia
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - Henry A Paz
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Ying Zhong
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Thunder Jalili
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
2
|
Abavisani M, Tafti P, Khoshroo N, Ebadpour N, Khoshrou A, Kesharwani P, Sahebkar A. The heart of the matter: How gut microbiota-targeted interventions influence cardiovascular diseases. Pathol Res Pract 2025; 269:155931. [PMID: 40174272 DOI: 10.1016/j.prp.2025.155931] [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: 09/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
The human body is habitat to a wide spectrum of microbial populations known as microbiota, which play an important role in overall health. The considerable research has mostly focused on the gut microbiota due to its potential to impact numerous physiological functions and its correlation with a variety of disorders, such as cardiovascular diseases (CVDs). Imbalances in the gut microbiota, known as dysbiosis, have been linked to the development and progression of CVDs through various processes, including the generation of metabolites like trimethylamine-N-oxide and short-chain fatty acids. Studies have also looked at the idea of using therapeutic interventions, like changing your diet, taking probiotics or prebiotics, or even fecal microbiota transplantation (FMT), to change the gut microbiota's make-up and how it works in order to prevent or treat CVDs. Exploring the cause-and-effect connection between the gut microbiota and CVDs offers a hopeful path for creating innovative microbiome-centered strategies to prevent and cure CVDs. This review presents an in-depth review of the correlation between the gut microbiota and CVDs, as well as potential therapeutic approaches for manipulating the gut microbiota to enhance cardiovascular health.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pourya Tafti
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khoshroo
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pardesh, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Slöcker-Barrio M, López-Herce Cid J, Solana-García MJ. The Interplay Between Nutrition and Microbiota and the Role of Probiotics and Symbiotics in Pediatric Infectious Diseases. Nutrients 2025; 17:1222. [PMID: 40218980 PMCID: PMC11990912 DOI: 10.3390/nu17071222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The interplay between nutrition and infectious diseases has been a central theme in health sciences for the last decades due to its great impact on the pediatric population, especially in immunocompromised patients and critically ill children. As conventional treatment and the development of antimicrobials for most infections standard treatment is either limited or not possible, alternative treatment options should be explored. Recent research shows that early enteral nutrition and nutritional supplements (such as probiotics and symbiotics) could have a pivotal role in promoting a healthy microbiome and subsequently preventing and improving outcomes for certain pediatric infectious diseases. However, understanding the specific mechanism of action and tailoring nutritional interventions remains a significant challenge. The optimal dose range for different probiotic strains and prebiotics and the most effective combination for each treatment indication needs further investigation and is yet to be defined. Additionally, in the era of personalized medicine, goal- and patient-directed treatment are key to optimizing and improving outcomes and minimizing potential complications and side effects, especially in complex and immunocompromised patients. The main objectives of this narrative review are 1. to explore the relationship and the complex interactions between microbiota and the human immune system; 2. to describe the influence of nutrition on infectious diseases; 3. to evaluate the impact of supplementation with probiotics and symbiotics in the prevention and treatment of the most relevant infections in children; and 4. to identify knowledge gaps and potential research priorities regarding the use of these supplements in pediatric patients.
Collapse
Affiliation(s)
- María Slöcker-Barrio
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain; (J.L.-H.C.); (M.J.S.-G.)
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID], RD24/0013/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gregorio Marañón Biomedical Research Institute, 28009 Madrid, Spain
| | - Jesús López-Herce Cid
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain; (J.L.-H.C.); (M.J.S.-G.)
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID], RD24/0013/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gregorio Marañón Biomedical Research Institute, 28009 Madrid, Spain
- Mother and Child and Public Health Department, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María José Solana-García
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain; (J.L.-H.C.); (M.J.S.-G.)
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID], RD24/0013/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gregorio Marañón Biomedical Research Institute, 28009 Madrid, Spain
- Mother and Child and Public Health Department, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
4
|
Zheng T, Meng C, Lv Z, Wu C, Zhou X, Mao W. The Critical Role of Faecalibacterium prausnitzii in Cardiovascular Diseases. Rev Cardiovasc Med 2025; 26:26740. [PMID: 40160596 PMCID: PMC11951488 DOI: 10.31083/rcm26740] [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] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
Due to the continued aging of the global population, cardiovascular diseases (CVDs) remain the main cause of death worldwide, with millions of fatalities from diseases, including stroke and coronary artery disease, reported annually. Thus, novel therapeutic approaches and targets are urgently required for diagnosing and treating CVDs. Recent studies emphasize the vital part of gut microbiota in both CVD prevention and management. Among these, Faecalibacterium prausnitzii (F. prausnitzii) has emerged as a promising probiotic capable of improving intestinal health. Although preliminary investigations demonstrate that F. prausnitzii positively enhances cardiovascular health, research specifically connecting this strain to CVD outcomes remains limited. Based on current research and assessment of possible clinical applications, this paper aimed to investigate the positive effects on cardiovascular health using F. prausnitzii and its metabolites. Targeting gut flora is expected to become a mainstay in CVD treatment as research develops.
Collapse
Affiliation(s)
- Tiantian Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenchen Meng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Zhengtian Lv
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenxia Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006 Hangzhou, Zhejiang, China
| | - Wei Mao
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, 310030 Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Hindle VK, Veasley NM, Holscher HD. Microbiota-Focused Dietary Approaches to Support Health: A Systematic Review. J Nutr 2025; 155:381-401. [PMID: 39486521 PMCID: PMC11867136 DOI: 10.1016/j.tjnut.2024.10.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Diet affects the intestinal microbiota. Increasingly, research is linking the intestinal microbiota to various human health outcomes. Consumption of traditional prebiotics (inulin, fructo-oligosaccharides, and galacto-oligosaccharides) confers health benefits through substrate utilization by select intestinal microorganisms, namely Bifidobacterium and Lactobacilli spp. A similar but distinct concept focused on microorganisms to support human health is through direct consumption of certain live microorganisms recognized as probiotics, which classically include Lactobacilli or Bifidobacterium strains. With advances in sequencing technologies and culturing techniques, other novel functional intestinal microorganisms are being increasingly identified and studied to determine how they may underpin human health benefits. These novel microorganisms are targeted for enrichment within the autochthonous intestinal microbiota through dietary approaches and are also gaining interest as next-generation probiotics because of their purported beneficial properties. Thus, characterizing dietary approaches that nourish select microorganisms in situ is necessary to propel biotic-focused research forward. As such, we reviewed the literature to summarize findings on dietary approaches that nourish the human intestinal microbiota and benefit health to help fill the gap in knowledge on the connections between certain microorganisms, the metabolome, and host physiology. The overall objective of this systematic review was to summarize the impact of dietary interventions with the propensity to nourish certain intestinal bacteria, affect microbial metabolite concentrations, and support gastrointestinal, metabolic, and cognitive health in healthy adults. Findings from the 17 randomized controlled studies identified in this systematic review indicated that dietary interventions providing dietary fibers, phytonutrients, or unsaturated fatty acids differentially enriched Akkermansia, Bacteroides, Clostridium, Eubacterium, Faecalibacterium, Roseburia, and Ruminococcus species, with variable effects on microbial metabolites and subsequent associations with physiologic markers of gastrointestinal and metabolic health. These findings have implications for biotic-focused research on candidate prebiotic substrates as well as next-generation probiotics.
Collapse
Affiliation(s)
- Veronica K Hindle
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Nadine M Veasley
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States; Personalized Nutrition Initiative, University of Illinois Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
6
|
Cai Y, Tang H, Xiang G, Yi H, Zhong J, Xie Z, Hu Q, El Bouhi R, Zhou P, Zhang Y, Yan H. Deciphering of differences in gut microbiota and plasma metabolites profile between non-obese and obese Golden Retrievers dogs. Front Microbiol 2025; 15:1514633. [PMID: 39845032 PMCID: PMC11751222 DOI: 10.3389/fmicb.2024.1514633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Golden Retrievers have a high risk of obesity, which is prevalent in dogs and is associated with inflammation and cancer, impairing the health and life expectancy of companion animals. Microbial and metabolite biomarkers have been proposed for identifying the presence of obesity in humans and rodents. However, the effects of obesity on the microbiome and metabolome of Golden Retrievers remains unknown. Therefore, this study was designed to evaluate the signatures of serum biochemistry indexes, gut microbiota and plasma metabolites in non-obese and obese Golden Retrievers, aiming to recognize potential biomarkers of canine obesity. Methods A total of 8 non-obese (Ctrl group) and 8 obese (Obe group) Golden Retrievers were included in the present study to collect blood and feces samples for measurements. The fecal microbiome and plasma metabolome were determined using 16S rRNA amplicon sequencing and liquid chromatography-mass spectrometry, respectively. Results Results showed that the alanine aminotransferase activity and total bilirubin concentration, which have been measured using serum biochemistry analysis, were higher in the Obe group than in the Ctrl group (p < 0.05). Moreover, there was a significant difference in gut microbiota composition between the two groups (p < 0.05). The phyla Proteobacteria, Fusobacteriota, and Bacteroidota as well as genera Fusobacterium, Prevotella, Faecalibacterium, Escherichia-Shigell, and Alloprevotella were more abundant, while phylum Firmicutes and genera Peptoclostridium, Blautia, Turicibacter, Allobaculum, and Erysipelatoclostridium were less abundant in the Obe group compared to the Ctrl group (p < 0.05). Plasma concentrations of citrulline and 11-dehydrocorticosterone were significantly higher in the Obe group than those in the Ctrl group (p < 0.05). Close correlations between serum biochemistry parameters, gut microbiome, and plasma metabolites were observed in the current study. Conclusion The obesity-induced shifts in serum biochemistry indexes, gut microbiota, and plasma metabolites profiles suggest that obese Golden Retrievers exhibit a different microbiome and metabolome than non-obese ones, and the certain metabolites like citrulline and 11-dehydrocorticosterone could be considered as potential biomarkers to recognize obese Golden Retrievers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Honglin Yan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| |
Collapse
|
7
|
Du J, Zhao X, Ding X, Han Q, Duan Y, Ren Q, Wang H, Song C, Wang X, Zhang D, Zhu H. The Role of the Gut Microbiota in Complications among Hemodialysis Patients. Microorganisms 2024; 12:1878. [PMID: 39338552 PMCID: PMC11434415 DOI: 10.3390/microorganisms12091878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
The composition of the gut microbiota varies among end-stage renal disease (ESRD) patients on the basis of their mode of renal replacement therapy (RRT), with notably more pronounced dysbiosis occurring in those undergoing hemodialysis (HD). Interventions such as dialysis catheters, unstable hemodynamics, strict dietary restrictions, and pharmacotherapy significantly alter the intestinal microenvironment, thus disrupting the gut microbiota composition in HD patients. The gut microbiota may influence HD-related complications, including cardiovascular disease (CVD), infections, anemia, and malnutrition, through mechanisms such as bacterial translocation, immune regulation, and the production of gut microbial metabolites, thereby affecting both the quality of life and the prognosis of patients. This review focuses on alterations in the gut microbiota and its metabolites in HD patients. Additionally, understanding the impact of the gut microbiota on the complications of HD could provide insights into the development of novel treatment strategies to prevent or alleviate complications in HD patients.
Collapse
Affiliation(s)
- Junxia Du
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaolin Zhao
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yingjie Duan
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Qinqin Ren
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Chenwen Song
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Dong Zhang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| |
Collapse
|
8
|
Yang X, Liu D, Zhao X, Han Y, Zhang X, Zhou Q, Lv Q. Hyperuricemia drives intestinal barrier dysfunction by regulating gut microbiota. Heliyon 2024; 10:e36024. [PMID: 39224259 PMCID: PMC11367111 DOI: 10.1016/j.heliyon.2024.e36024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Background Hyperuricemia elevates gut permeability; however, the risk of its influence on the compromised intestinal barrier is poorly understood. Aims This study was carried out, aiming to elucidate the orchestrators and disruptors of intestinal barrier in hyperuricemia. Methods A mouse model of hyperuricemia was induced by administering adenine and oteracil potassium to mice. Allopurinol was used to decrease uric acid level, and antibiotics were administered to mice to deplete gut microbiota. Intestinal permeability was assessed using FITC-labeled dextran. Changes in gut microbial community were analyzed through 16S rRNA sequencing. IL-1β and TNF-α levels were quantified using ELISA. The expression of tight junction protein genes, TLR4, p65 and IL-1β, was determined with Q-PCR and Western blotting. Results Allopurinol treatment effectively reduced intestinal permeability and serum TNF-α levels. Antibiotic treatment alleviated but not abolished intestinal permeability. Uric acid alone was insufficient to increase Coca2 monolayer permeability. Allopurinol treatment altered microbial composition and suppressed opportunistic infections. Re-establishing hyperuricemia in a germfree mouse model protected mice from intestinal injury. Allopurinol and antibiotic treatments reduced TLR4 and IL-1β expressions, increased occludin and claudin-1 expressions but suppressed NF-ĸB p65 signaling. However, removing gut microbiota aggravated lipid metabolic dysfunction. Conclusion Gut microbiota is a direct and specific cause for intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Xiaomin Yang
- Laboratory Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Dan Liu
- Laboratory Medicine, Qingdao Fuwai Cardiovascular Hospital, PR China
| | - Xiangzhong Zhao
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Yafei Han
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Xiao Zhang
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Quan Zhou
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Qiulan Lv
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| |
Collapse
|
9
|
Randeni N, Bordiga M, Xu B. A Comprehensive Review of the Triangular Relationship among Diet-Gut Microbiota-Inflammation. Int J Mol Sci 2024; 25:9366. [PMID: 39273314 PMCID: PMC11394685 DOI: 10.3390/ijms25179366] [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: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
The human gastrointestinal tract hosts a complex and dynamic community of microorganisms known as the gut microbiota, which play a pivotal role in numerous physiological processes, including digestion, metabolism, and immune function. Recent research has highlighted the significant impact of diet on the gut microbiota composition and functionality, and the consequential effects on host health. Concurrently, there is growing evidence linking the gut microbiota to inflammation, a key factor in many chronic diseases such as inflammatory bowel disease (IBD), obesity, diabetes, and cardiovascular diseases (CVDs). This review explores how dietary components influence the gut microbiota composition, how these microbial changes affect inflammatory pathways, and the therapeutic implications of modulating this axis for chronic inflammatory disease prevention and management. Beneficial dietary patterns, such as the Mediterranean diet (MD) and plant-based diets, promote a diverse and balanced gut microbiota composition, supporting anti-inflammatory pathways. Conversely, the Western diet (WD), high in saturated fats and refined sugars, is associated with dysbiosis and increased inflammation. With all the links between the three variables considered, this review attempts to offer a thorough examination of the triangle formed by inflammation, the gut microbiota, and food.
Collapse
Affiliation(s)
- Nidesha Randeni
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
- Department of Agricultural and Plantation Engineering, Faculty of Engineering Technology, The Open University of Sri Lanka, Nawala, Nugegoda 10250, Sri Lanka
| | - Matteo Bordiga
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100 Novara, Italy
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai 519087, China
| |
Collapse
|
10
|
I Fernández-Avila A, Gutiérrez-Ibanes E, Martín de Miguel I, Sanz-Ruiz R, Gabaldón Á, Fernández-Avilés F, Gómez-Lara J, Fernández-Castillo M, Vázquez-Cuesta S, Martínez-Legazpi P, Lozano-Garcia N, Blázquez-López E, Yotti R, López-Cade I, Reigadas E, Muñoz P, Elízaga J, Correa R, Bermejo J. One-year longitudinal changes of peripheral CD4+ T-lymphocyte counts, gut microbiome, and plaque vulnerability after an acute coronary syndrome. IJC HEART & VASCULATURE 2024; 53:101438. [PMID: 38912228 PMCID: PMC11190720 DOI: 10.1016/j.ijcha.2024.101438] [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: 03/01/2024] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024]
Abstract
Background Longitudinal changes in gut microbiome and inflammation may be involved in the evolution of atherosclerosis after an acute coronary syndrome (ACS). We aimed to characterize repeated profiles of gut microbiota and peripheral CD4+ T lymphocytes during the first year after an ACS, and to address their relationship with atherosclerotic plaque changes. Methods Over one year we measured the microbiome, peripheral counts of CD4+ T populations and cytokines in 67 patients shortly after a first ACS. We compared baseline measurements to those of a matched population of 40 chronic patients. A subgroup of 20 ACS patients underwent repeated assessment of fibrous cap thickness (FCT) of a non-culprit lesion. Results At admission, ACS patients showed gut dysbiosis compared with the chronic group, which was rapidly reduced and remained low at 1-year. Also, their Th1 and Th2 CD4+ T counts were increased but decreased over time. The CD4+ T counts were related to ongoing changes in gut microbiome. Unsupervised clustering of repeated CD4+ Th0, Th1, Th2, Th17 and Treg counts in ACS patients identified two different cell trajectory patterns, related to cytokines. The group of patients following a high-CD4+ T cell trajectory showed a one-year reduction in their FCT [net effect = -24.2 µm; p = 0.016]. Conclusions Patients suffering an ACS show altered profiles of microbiome and systemic inflammation that tend to mimic values of chronic patients after 1-year. However, in one-third of patients, this inflammatory state remains particularly dysregulated. This persistent inflammation is likely related to plaque vulnerability as evident by fibrous cap thinning (Clinical Trial NCT03434483).
Collapse
Affiliation(s)
- Ana I Fernández-Avila
- 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
| | - Irene Martín de Miguel
- 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
| | - Ricardo Sanz-Ruiz
- 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
| | - Álvaro Gabaldón
- 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
| | - 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
| | - Josep Gómez-Lara
- Department of Cardiology, Hospital Universitario de Bellvitge, and CIBERCV, Barcelona, Spain
| | - Marta Fernández-Castillo
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Silvia Vázquez-Cuesta
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERES, Madrid, Spain
| | - Pablo Martínez-Legazpi
- 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
- Department of Mathematical Physics and Fluids, Facultad de Ciencias, Universidad Nacional de Educación a Distancia, UNED, Madrid, Spain
| | - Nuria Lozano-Garcia
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERES, Madrid, Spain
| | - Elena Blázquez-López
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón, 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
| | - Igor López-Cade
- 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
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERES, Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERES, Madrid, Spain
| | - Jaime Elízaga
- 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
| | - Rafael Correa
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón, 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
| |
Collapse
|
11
|
Zhang H, Zhou Y, Pan Z, Wang B, Yang L, Zhang N, Chen B, Wang X, Jian Z, Wang L, Ling H, Qin X, Zhang Z, Liu T, Zheng A, Tan Y, Bi Y, Yang R. Toxicity assessment of Cucurbita pepo cv Dayangua and its effects on gut microbiota in mice. BMC Complement Med Ther 2024; 24:243. [PMID: 38909225 PMCID: PMC11193904 DOI: 10.1186/s12906-024-04551-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 06/12/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Cucurbita pepo cv Dayangua (CPD) is an edible plant with diverse pharmacological properties. The current research on CPD has primarily focused on initial investigations of its chemical composition and pharmacological effects, and no comprehensive toxicity assessment has been conducted to date. METHODS In the present study, the toxicity of CPD was evaluated through both acute and sub-chronic oral toxicity tests in mice. 16S rDNA sequencing was used to analyze the composition of the gut microbiota of mice at different time points to observe the effect of CPD on these microbial communities. RESULTS In the acute toxicity test, CPD exhibited low toxicity, with a median lethal dose (LD50) > 2000 mg/kg. The sub-chronic toxicity test indicated that CPD administration at doses of 200, 400, and 600 mg/kg did not cause mortality or significant organ damage in mice. Furthermore, analysis of the gut microbiota after gavage administration of CPD at 400 and 600 mg/kg revealed an improved abundance of some beneficial gut bacteria. CONCLUSIONS In summary, no acute or sub-chronic toxic effects were observed in mice following the oral administration of CPD. CPD did not affect the structure and diversity of the gut microbiota and may contribute to an increase in the number of beneficial gut bacteria.
Collapse
Affiliation(s)
- Huan Zhang
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yazhou Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Bikun Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Lei Yang
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
| | - Nan Zhang
- Heilongjiang Biodi Bio-Pharma Technology Company Lmt., No. 178, Yuexiujie, Harbin, Heilongjiang Province, China
| | - Baiyi Chen
- Heilongjiang Biodi Bio-Pharma Technology Company Lmt., No. 178, Yuexiujie, Harbin, Heilongjiang Province, China
| | - Xiaona Wang
- Heilongjiang Biodi Bio-Pharma Technology Company Lmt., No. 178, Yuexiujie, Harbin, Heilongjiang Province, China
| | - Zhiguang Jian
- Heilongjiang Biodi Bio-Pharma Technology Company Lmt., No. 178, Yuexiujie, Harbin, Heilongjiang Province, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Hui Ling
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Xiaoming Qin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Zhelin Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Teng Liu
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Aiping Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yafang Tan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Ruifu Yang
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China.
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China.
| |
Collapse
|
12
|
Bridge LA, Hernández Vargas JA, Trujillo-Cáceres SJ, Beigrezaei S, Chatelan A, Salehi-Abargouei A, Muka T, Uriza-Pinzón JP, Raeisi-Dehkordi H, Franco OH, Grompone G, Artola Arita V. Two cosmoses, one universe: a narrative review exploring the gut microbiome's role in the effect of urban risk factors on vascular ageing. Maturitas 2024; 184:107951. [PMID: 38471294 DOI: 10.1016/j.maturitas.2024.107951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024]
Abstract
In the face of rising global urbanisation, understanding how the associated environment and lifestyle impact public health is a cornerstone for prevention, research, and clinical practice. Cardiovascular disease is the leading cause of morbidity and mortality worldwide, with urban risk factors contributing greatly to its burden. The current narrative review adopts an exposome approach to explore the effect of urban-associated physical-chemical factors (such as air pollution) and lifestyle on cardiovascular health and ageing. In addition, we provide new insights into how these urban-related factors alter the gut microbiome, which has been associated with an increased risk of cardiovascular disease. We focus on vascular ageing, before disease onset, to promote preventative research and practice. We also discuss how urban ecosystems and social factors may interact with these pathways and provide suggestions for future research, precision prevention and management of vascular ageing. Most importantly, future research and decision-making would benefit from adopting an exposome approach and acknowledging the diverse and boundless universe of the microbiome.
Collapse
Affiliation(s)
- Lara Anne Bridge
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Juliana Alexandra Hernández Vargas
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Silvia Juliana Trujillo-Cáceres
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sara Beigrezaei
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Angeline Chatelan
- Geneva School of Health Sciences, HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Amin Salehi-Abargouei
- Research Center for Food Hygiene and Safety, Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Julieth Pilar Uriza-Pinzón
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hamidreza Raeisi-Dehkordi
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Oscar H Franco
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Vicente Artola Arita
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
13
|
Yang Y, Liu X, Liu X, Xie C, Shi J. The role of the kynurenine pathway in cardiovascular disease. Front Cardiovasc Med 2024; 11:1406856. [PMID: 38883986 PMCID: PMC11176437 DOI: 10.3389/fcvm.2024.1406856] [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: 03/25/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
The kynurenine pathway (KP) serves as the primary route for tryptophan metabolism in most mammalian organisms, with its downstream metabolites actively involved in various physiological and pathological processes. Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) serve as the initial and pivotal enzymes of the KP, with IDO playing important and intricate roles in cardiovascular diseases. Multiple metabolites of KP have been observed to exhibit elevated concentrations in plasma across various cardiovascular diseases, such as atherosclerosis, hypertension, and acute myocardial infarction. Multiple studies have indicated that kynurenine (KYN) may serve as a potential biomarker for several adverse cardiovascular events. Furthermore, Kynurenine and its downstream metabolites have complex roles in inflammation, exhibiting both inhibitory and stimulatory effects on inflammatory responses under different conditions. In atherosclerosis, upregulation of IDO stimulates KYN production, mediating aromatic hydrocarbon receptor (AhR)-induced exacerbation of vascular inflammation and promotion of foam cell formation. Conversely, in arterial calcification, this mediation alleviates osteogenic differentiation of vascular smooth muscle cells. Additionally, in cardiac remodeling, KYN-mediated AhR activation exacerbates pathological left ventricular hypertrophy and fibrosis. Interventions targeting components of the KP, such as IDO inhibitors, 3-hydroxyanthranilic acid, and anthranilic acid, demonstrate cardiovascular protective effects. This review outlines the mechanistic roles of KP in coronary atherosclerosis, arterial calcification, and myocardial diseases, highlighting the potential diagnostic, prognostic, and therapeutic value of KP in cardiovascular diseases, thus providing novel insights for the development and application of related drugs in future research.
Collapse
Affiliation(s)
- Yuehang Yang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chiyang Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Wang H, Wang Y, Yang L, Feng J, Tian S, Chen L, Huang W, Liu J, Wang X. Integrated 16S rRNA sequencing and metagenomics insights into microbial dysbiosis and distinct virulence factors in inflammatory bowel disease. Front Microbiol 2024; 15:1375804. [PMID: 38591039 PMCID: PMC10999624 DOI: 10.3389/fmicb.2024.1375804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/26/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction The escalation of urbanization correlates with rising rates of inflammatory bowel disease (IBD), necessitating research into new etiological factors. This study aims to elucidate the gut microbiota profiles in IBD patients and compare them with healthy controls in a western city of China. Methods We conducted a multicenter case-control study from the end of 2020, using 16S rRNA gene sequencing (n = 36) and metagenomic sequencing (n = 12) to analyze the gut microbiota of newly diagnosed IBD patients, including those with Crohn's disease (CD) and ulcerative colitis (UC). Results Our results demonstrated a significant enrichment of the phylum Proteobacteria, particularly the genus Escherichia-Shigella, in CD patients. Conversely, the genus Enterococcus was markedly increased in UC patients. The core gut microbiota, such as the Christensenellaceae R-7 group, Fusicatenibacter, and Holdemanella, were primarily identified in healthy subjects. Additionally, significant interactions between the microbiome and virulence factors were observed. Discussion The findings suggest that oxidative stress may play a pivotal role in the pathology of IBD. This study contributes to the growing dialogue about the impact of gut microbiota on the development of IBD and its variations across different geographies, highlighting potential avenues for further research.
Collapse
Affiliation(s)
- Haijing Wang
- Medical College of Qinghai University, Xining, China
| | - Yuanjun Wang
- Medical College of Qinghai University, Xining, China
- Qinghai University Affiliated Hospital, Xining, China
| | - Libin Yang
- Ningxia Hui Autonomous Region People's Hospital, Yinchuan, China
| | - Jiawen Feng
- Medical College of Qinghai University, Xining, China
- Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| | - Shou Tian
- Medical College of Qinghai University, Xining, China
- Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| | - Lingyan Chen
- Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| | - Wei Huang
- Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| | - Jia Liu
- Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| | - Xiaojin Wang
- Medical College of Qinghai University, Xining, China
- Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| |
Collapse
|
15
|
Langhi C, Vallier M, Bron A, Otero YF, Maura M, Le Joubioux F, Blomberg N, Giera M, Guigas B, Maugard T, Chassaing B, Peltier S, Blanquet-Diot S, Bard JM, Sirvent P. A polyphenol-rich plant extract prevents hypercholesterolemia and modulates gut microbiota in western diet-fed mice. Front Cardiovasc Med 2024; 11:1342388. [PMID: 38317864 PMCID: PMC10839041 DOI: 10.3389/fcvm.2024.1342388] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Totum-070 is a combination of five plant extracts enriched in polyphenols to target hypercholesterolemia, one of the main risk factors for cardiovascular diseases. The aim of this study was to investigate the effects of Totum-070 on cholesterol levels in an animal model of diet-induced hypercholesterolemia. Methods C57BL/6JOlaHsd male mice were fed a Western diet and received Totum-070, or not, by daily gavage (1g/kg and 3g/kg body weight) for 6 weeks. Results The Western diet induced obesity, fat accumulation, hepatic steatosis and increased plasma cholesterol compared with the control group. All these metabolic perturbations were alleviated by Totum-070 supplementation in a dose-dependent manner. Lipid excretion in feces was higher in mice supplemented with Totum-070, suggesting inhibition of intestinal lipid absorption. Totum-070 also increased the fecal concentration of short chain fatty acids, demonstrating a direct effect on intestinal microbiota. Discussion The characterization of fecal microbiota by 16S amplicon sequencing showed that Totum-070 supplementation modulated the dysbiosis associated with metabolic disorders. Specifically, Totum-070 increased the relative abundance of Muribaculum (a beneficial bacterium) and reduced that of Lactococcus (a genus positively correlated with increased plasma cholesterol level). Together, these findings indicate that the cholesterol-lowering effect of Totum-070 bioactive molecules could be mediated through multiple actions on the intestine and gut microbiota.
Collapse
Affiliation(s)
| | | | - Auriane Bron
- UMR 454 Microbiologie Environnement DIgestif et Santé (MEDIS), Université Clermont Auvergne, Clermont-Ferrand, France
| | | | | | | | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Thierry Maugard
- Equipe BCBS (Biotechnologies et Chimie des Bioressources Pour la Santé), UMR CNRS 7266 LIENSs, La Rochelle Université, La Rochelle, France
| | - Benoit Chassaing
- Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris, France
| | | | - Stéphanie Blanquet-Diot
- UMR 454 Microbiologie Environnement DIgestif et Santé (MEDIS), Université Clermont Auvergne, Clermont-Ferrand, France
| | - Jean-Marie Bard
- Laboratoire de Biochimie Générale et Appliquée, UFR de Pharmacie, ISOMer-UE 2160, IUML-Institut Universitaire Mer et Littoral-FR3473 CNRS, Université de Nantes, Nantes, France
| | | |
Collapse
|
16
|
Zhong Q, Reyes-Jurado F, Calumba KF. Structured soft particulate matters for delivery of bioactive compounds in foods and functioning in the colon. SOFT MATTER 2024; 20:277-293. [PMID: 38090993 DOI: 10.1039/d3sm00866e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
The present review discusses challenges, perspectives, and current needs of delivering bioactive compounds (BCs) using soft particulate matters (SPMs) for gut health. SPMs can entrap BCs for incorporation in foods, preserve their bioactivities during processing, storage, and gastrointestinal digestion, and deliver BCs to functioning sites in the colon. To enable these functions, physical, chemical, and biological properties of BCs are integrated in designing various types of SPMs to overcome environmental factors reducing the bioavailability and bioactivity of BCs. The design principles are applied using food grade molecules with the desired properties to produce SPMs by additionally considering the cost, sustainability, and scalability of manufacturing processes. Lastly, to make delivery systems practical, impacts of SPMs on food quality are to be evaluated case by case, and health benefits of functional foods incorporated with delivery systems are to be confirmed and must outweigh the cost of preparing SPMs.
Collapse
Affiliation(s)
- Qixin Zhong
- Department of Food Science, University of Tennessee, Knoxville, TN, USA.
| | | | - Kriza Faye Calumba
- Department of Food Science, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
17
|
Usman I, Anwar A, Shukla S, Pathak P. Mechanistic Review on the Role of Gut Microbiota in the Pathology of Cardiovascular Diseases. Cardiovasc Hematol Disord Drug Targets 2024; 24:13-39. [PMID: 38879769 DOI: 10.2174/011871529x310857240607103028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 07/31/2024]
Abstract
Cardiovascular diseases (CVDs), which stand as the primary contributors to illness and death on a global scale, include vital risk factors like hyperlipidemia, hypertension, diabetes, and smoking, to name a few. However, conventional cardiovascular risk factors offer only partial insight into the complexity of CVDs. Lately, a growing body of research has illuminated that the gut microbiome and its by-products are also of paramount importance in the initiation and progression of CVDs. The gastrointestinal tract houses trillions of microorganisms, commonly known as gut microbiota, that metabolize nutrients, yielding substances like trimethylamine-N-oxide (TMAO), bile acids (BAs), short-chain fatty acids (SCFAs), indoxyl sulfate (IS), and so on. Strategies aimed at addressing these microbes and their correlated biological pathways have shown promise in the management and diagnosis of CVDs. This review offers a comprehensive examination of how the gut microbiota contributes to the pathogenesis of CVDs, particularly atherosclerosis, hypertension, heart failure (HF), and atrial fibrillation (AF), explores potential underlying mechanisms, and highlights emerging therapeutic prospects in this dynamic domain.
Collapse
Affiliation(s)
- Iqra Usman
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Aamir Anwar
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Shivang Shukla
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| | - Priya Pathak
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Lucknow Campus, U.P., 226010, India
| |
Collapse
|
18
|
Moriki D, Koumpagioti D, Francino MP, Rufián-Henares JÁ, Kalogiannis M, Priftis KN, Douros K. How Different Are the Influences of Mediterranean and Japanese Diets on the Gut Microbiome? Endocr Metab Immune Disord Drug Targets 2024; 24:1733-1745. [PMID: 38243975 DOI: 10.2174/0118715303261069231124092259] [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: 05/06/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 01/22/2024]
Abstract
The gut microbiome is a complex ecosystem, mainly composed of bacteria, that performs essential functions for the host. Its composition is determined by many factors; however, diet has emerged as a key regulator. Both the Mediterranean (MD) and Japanese (JD) diets have been associated with significant health benefits and are therefore considered healthy dietary patterns. Both are plant-based diets and although they have much in common, they also have important differences mainly related to total calorie intake and the consumption of specific foods and beverages. Thus, it has been hypothesized that they exert their beneficial properties through different nutrients and bioactive compounds that interact with gut microbes and induce specific changes on gut metabolic pathways. In this review, we present current data on the effects of the MD and JD on the gut microbiome. Furthermore, we aim to examine whether there are differences or shared effects on the gut microbiome of people who adhere to these dietary patterns.
Collapse
Affiliation(s)
- Dafni Moriki
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Despoina Koumpagioti
- Department of Nursing, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Pilar Francino
- Department of Genomics and Health, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
- CIBER en Epidemiología y Salud Pública, 28029 Madrid, Spain
| | - José Ángel Rufián-Henares
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica, Universidad de Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Universidad de Granada, 18071 Granada, Spain
| | - Michalis Kalogiannis
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Kostas N Priftis
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Konstantinos Douros
- Allergology and Pulmonology Unit, 3rd Pediatric Department, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
19
|
Dinu LD, Gatea F, Matei F, Banciu C, Vamanu E. Gut Microbiota Modulation by Selenium and Zinc Enrichment Postbiotic on Dysbiosis Associated with Hypertension. Curr Vasc Pharmacol 2024; 22:365-374. [PMID: 38779729 DOI: 10.2174/0115701611290537240509061549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Targeting gut dysbiosis to treat chronic diseases or to alleviate the symptoms is a new direction for medical adjuvant therapies. Recently, postbiotics have received considerable attention as they are non-viable probiotic preparations that confer various health benefits to the host without the safety problems associated with using live microbial cells. OBJECTIVE The aim of the study is to obtain selenium (Se) and zinc (Zn) enriched Saccharomyces boulardii postbiotic biomass and to analyze its modulation effect because these minerals play an important role in reducing gut dysbiosis linked to cardiovascular (CV) diseases. METHOD The effect of the S. boulardii and Se/Zn enriched yeast postbiotics on CV microbial fingerprint was studied in vitro using the gastrointestinal system (GIS 1) and analyzed by microbiological, chemical, and qPCR methods. RESULT There was a 2.2 log CFU/mL increase in the total bacterial load after SeZn postbiotic treatment and in the qPCR counts of Firmicutes phyla for both treatments. Beneficial taxa, Bifidobacterium spp. and Lactobacillus spp., as well as Bacteroidesspp. were up to 1.5 log higher after mineral- enriched postbiotic application, while the acetic acid level increased. CONCLUSION These preliminary studies highlight the therapeutic potential of using Se/Zn enriched yeast postbiotics as adjuvants for clinical treatments of CV diseases.
Collapse
Affiliation(s)
- Laura-Dorina Dinu
- Department of Industrial Biotechnology, Faculty of Biotechnology, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| | - Florentina Gatea
- Department of Biotechnology, Centre of Bioanalysis, National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Florentina Matei
- Faculty of Food Industry and Tourism, Transylvania University of Brasov, Brasov, Romania
| | - Cristian Banciu
- Department of Ecology, Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Emanuel Vamanu
- Department of Industrial Biotechnology, Faculty of Biotechnology, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| |
Collapse
|
20
|
Hamjane N, Mechita MB, Nourouti NG, Barakat A. Gut microbiota dysbiosis -associated obesity and its involvement in cardiovascular diseases and type 2 diabetes. A systematic review. Microvasc Res 2024; 151:104601. [PMID: 37690507 DOI: 10.1016/j.mvr.2023.104601] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/24/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Obesity is a complex, multifactorial disease caused by various factors. Recently, the role of the gut microbiota in the development of obesity and its complications has attracted increasing interest. PURPOSE This article focuses on the mechanisms by which gut microbiota dysbiosis induces insulin resistance, type 2 diabetes, and cardiovascular diseases linked to obesity, highlighting the mechanisms explaining the role of gut microbiota dysbiosis-associated inflammation in the onset of these pathologies. METHODS A systematic study was carried out to understand and summarize the published results on this topic. More than 150 articles were included in this search, including different types of studies, consulted by an online search in English using various electronic search databases and predefined keywords related to the objectives of our study. RESULTS We have summarized the data from the articles consulted in this search, and we have found a major gut microbiota alteration in obesity, characterized by a specific decrease in butyrate-producing bacteria and the production of metabolites and components that lead to metabolic impairments and affect the progression of various diseases associated with obesity through distinct signaling pathways, including insulin resistance, type 2 diabetes, and cardiovascular diseases (CVD). We have also focused on the major role of inflammation as a link between gut microbiota dysbiosis and obesity-associated metabolic complications by explaining the mechanisms involved. CONCLUSION Gut microbiota dysbiosis plays a crucial role in the development of various obesity-related metabolic abnormalities, among them type 2 diabetes and CVD, and represents a major challenge for chronic disease prevention and health. Indeed, the intestinal microbiota appears to be a promising target for the nutritional or therapeutic management of these diseases.
Collapse
Affiliation(s)
- Nadia Hamjane
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco.
| | - Mohcine Bennani Mechita
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco
| | - Naima Ghailani Nourouti
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco
| | - Amina Barakat
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco
| |
Collapse
|
21
|
Langhi C, Vallier M, Otero YF, Maura M, Le Joubioux F, Groult H, Achour O, Pebriana RB, Giera M, Guigas B, Maugard T, Chassaing B, Peltier S, Bard JM, Sirvent P. Totum-070, a Polyphenol-Rich Plant Extract, Prevents Hypercholesterolemia in High-Fat Diet-Fed Hamsters by Inhibiting Intestinal Cholesterol Absorption. Nutrients 2023; 15:5056. [PMID: 38140315 PMCID: PMC10746001 DOI: 10.3390/nu15245056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of mortality worldwide, and hypercholesterolemia is a central risk factor for atherosclerosis. This study evaluated the effects of Totum-070, a plant-based polyphenol-rich supplement, in hamsters with high-fat diet (HFD)-induced dyslipidemia. The molecular mechanisms of action were explored using human Caco2 enterocytes. Totum-070 supplementation reduced the total cholesterol (-41%), non-HDL cholesterol (-47%), and triglycerides (-46%) in a dose-dependent manner, compared with HFD. HFD-induced hepatic steatosis was also significantly decreased by Totum-070, an effect associated with the reduction in various lipid and inflammatory gene expression. Upon challenging with olive oil gavage, the post-prandial triglyceride levels were strongly reduced. The sterol excretion in the feces was increased in the HFD-Totum-070 groups compared with the HFD group and associated with reduction of intestinal cholesterol absorption. These effects were confirmed in the Caco2 cells, where incubation with Totum-070 inhibited cholesterol uptake and apolipoprotein B secretion. Furthermore, a microbiota composition analysis revealed a strong effect of Totum-070 on the alpha and beta diversity of bacterial species and a significant decrease in the Firmicutes to Bacteroidetes ratio. Altogether, our findings indicate that Totum-070 lowers hypercholesterolemia by reducing intestinal cholesterol absorption, suggesting that its use as dietary supplement may be explored as a new preventive strategy for cardiovascular diseases.
Collapse
Affiliation(s)
- Cédric Langhi
- R&D Riom Center, Valbiotis, 20-22 rue Henri et Gilberte Goudier, 63200 Riom, France
| | - Marie Vallier
- R&D Riom Center, Valbiotis, 20-22 rue Henri et Gilberte Goudier, 63200 Riom, France
| | - Yolanda F. Otero
- R&D Riom Center, Valbiotis, 20-22 rue Henri et Gilberte Goudier, 63200 Riom, France
| | - Maheva Maura
- R&D Center, Valbiotis, 23 Avenue Albert Einstein, 17000 La Rochelle, France
| | | | - Hugo Groult
- Equipe BCBS (Biotechnologies et Chimie des Bioressources pour la Santé), UMR (Unité Mixte de Recherche) CNRS (Centre National de la Recherche Scientifique) 7266 LIENSs (LIttoral ENvironnement Et Sociétés), La Rochelle Université, 17042 La Rochelle, France
| | - Oussama Achour
- BioAqtiv, Equipe BCBS (Biotechnologies et Chimie des Bioressources pour la Santé), LIENSs (LIttoral ENvironnement Et Sociétés), UMR (Unité Mixte de Recherche) 7266 CNRS (Centre National de la Recherche Scientifique), La Rochelle Université, 17042 La Rochelle, France
| | - Ratna Budhi Pebriana
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albi-nusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albi-nusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Thierry Maugard
- Equipe BCBS (Biotechnologies et Chimie des Bioressources pour la Santé), UMR (Unité Mixte de Recherche) CNRS (Centre National de la Recherche Scientifique) 7266 LIENSs (LIttoral ENvironnement Et Sociétés), La Rochelle Université, 17042 La Rochelle, France
| | - Benoit Chassaing
- Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, Institut Cochin, INSERM (Institut National de la Santé et de la Recherche Médicale) U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
| | - Sébastien Peltier
- R&D Périgny Center, Valbiotis, 12F rue Paul Vatine, 17180 Périgny, France
| | - Jean-Marie Bard
- Laboratoire de Biochimie Générale et Appliquée, UFR (Unité de Formation et de Recherche) de Pharmacie, ISOMer-UE 2160, IUML-Institut Universitaire Mer et Littoral-FR3473 CNRS, Université de Nantes, 44035 Nantes, France
| | - Pascal Sirvent
- R&D Riom Center, Valbiotis, 20-22 rue Henri et Gilberte Goudier, 63200 Riom, France
| |
Collapse
|
22
|
Ahmad AF, Caparrós-Martin JA, Gray N, Lodge S, Wist J, Lee S, O'Gara F, Shah A, Ward NC, Dwivedi G. Insights into the associations between the gut microbiome, its metabolites, and heart failure. Am J Physiol Heart Circ Physiol 2023; 325:H1325-H1336. [PMID: 37737730 DOI: 10.1152/ajpheart.00436.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/05/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
Heart failure (HF) is the end stage of most cardiovascular diseases and remains a significant health problem globally. We aimed to assess whether patients with left ventricular ejection fraction ≤45% had alterations in both the gut microbiome profile and production of associated metabolites when compared with a healthy cohort. We also examined the associated inflammatory, metabolomic, and lipidomic profiles of patients with HF. This single center, observational study, recruited 73 patients with HF and 59 healthy volunteers. Blood and stool samples were collected at baseline and 6-mo follow-up, along with anthropometric and clinical data. When compared with healthy controls, patients with HF had reduced gut bacterial alpha diversity at follow-up (P = 0.004) but not at baseline. The stool microbiota of patients with HF was characterized by a depletion of operational taxonomic units representing commensal Clostridia at both baseline and follow-up. Patients with HF also had significantly elevated baseline plasma acetate (P = 0.007), plasma trimethylamine-N-oxide (TMAO) (P = 0.003), serum soluble CD14 (sCD14; P = 0.005), and soluble CD163 (sCD163; P = 0.004) levels compared with healthy controls. Furthermore, patients with HF had a distinct metabolomic and lipidomic profile at baseline when compared with healthy controls. Differences in the composition of the gut microbiome and the levels of associated metabolites were observed in patients with HF when compared with a healthy cohort. This was also associated with an altered metabolomic and lipidomic profile. Our study identifies microorganisms and metabolites that could represent new therapeutic targets and diagnostic tools in the pathogenesis of HF.NEW & NOTEWORTHY We found a reduction in gut bacterial alpha diversity in patients with heart failure (HF) and that the stool microbiota of patients with HF was characterized by depletion of operational taxonomic units representing commensal Clostridia at both baseline and follow-up. Patients with HF also had altered bacterial metabolites and increased inflammatory profiles compared with healthy controls. A distinct metabolomic and lipidomic profile was present in patients with HF at baseline when compared with healthy controls.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Jose A Caparrós-Martin
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Microbiology, PathWest Laboratory Medicine, Perth, Western Australia, Australia
| | - Fergal O'Gara
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
| | - Amit Shah
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Natalie C Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
23
|
Kahleova H, Holtz DN, Strom N, La Reau A, Kolipaka S, Schmidt N, Hata E, Znayenko-Miller T, Holubkov R, Barnard ND. A dietary intervention for postmenopausal hot flashes: A potential role of gut microbiome. An exploratory analysis. Complement Ther Med 2023; 79:103002. [PMID: 37949415 DOI: 10.1016/j.ctim.2023.103002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/11/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE This study examined the role of gut microbiome changes in mediating the effects of a dietary intervention on the frequency and severity of postmenopausal vasomotor symptoms METHODS: Postmenopausal women (n = 84) reporting ≥2 moderate-to-severe hot flashes daily were randomly assigned, in 2 successive cohorts, to an intervention including a low-fat, vegan diet and cooked soybeans (½ cup [86 g] daily) or to stay on their usual diet. Over a 12-week period, frequency and severity of hot flashes were recorded with a mobile application. In a subset of 11 women, gut microbiome was analyzed at baseline and after 12 weeks of the dietary intervention (low-fat vegan diet with soybeans), using deep shotgun metagenomic sequencing. Differences in the microbiome between baseline and 12 weeks were assessed by comparing alpha diversity with Wilcoxon signed rank tests, beta diversity with permanovaFL, and taxon abundance with Wilcoxon signed rank tests. Pearson correlations were used to assess the association between changes in hot flashes and gut bacteria. RESULTS In the subset for which microbiome testing was done, total hot flashes decreased by 95 % during the dietary intervention (p = 0.007); severe hot flashes disappeared (from 0.6 to 0.0/day; p = 0.06); and moderate-to-severe hot flashes decreased by 96 % (p = 0.01). Daytime and nighttime hot flashes were reduced by 96 % (p = 0.01) and 94 % (p = 0.004), respectively. Alpha and beta diversity did not significantly differ in the intervention group between baseline and 12 weeks. Two families (Enterobacteriaceae and Veillonellaceae), 5 genera (Erysipelatoclostridium, Fusicatenibacter, Holdemanella, Intestinimonas, and Porphyromonas), and 6 species (Clostridium asparagiforme, Clostridium innocuum, Bacteroides thetaiotaomicron, Fusicatenibacter saccharivorans, Intestinimonas butyriciproducens, Prevotella corporis, and Streptococcus sp.) were differentially abundant, but after correction for multiple comparisons, these differences were no longer significant. Changes in the relative abundance of Porphyromonas and Prevotella corporis were associated with the reduction in severe day hot flashes both unadjusted (r = 0.61; p = 0.047; and r = 0.69; p = 0.02), respectively), and after adjustment for changes in body mass index (r = 0.63; p = 0.049; and r = 0.73; p = 0.02), respectively). Changes in relative abundance of Clostridium asparagiforme were associated with the reduction in total severe hot flashes (r = 0.69; p = 0.019) and severe night hot flashes (r = 0.82; p = 0.002) and the latter association remained significant after adjustment for changes in body mass index (r = 0.75; p = 0.01). CONCLUSIONS This exploratory analysis revealed potential associations between changes in vasomotor symptoms in response to a diet change and changes in the gut microbiome. Larger randomized clinical trials are needed to investigate these findings.
Collapse
Affiliation(s)
- Hana Kahleova
- Physicians Committee for Responsible Medicine, Washington, DC, USA.
| | - Danielle N Holtz
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | | | | | - Sinjana Kolipaka
- Physicians Committee for Responsible Medicine, Washington, DC, USA; Florida Atlantic University, Charles E. Schmidt College of Medicine, FL, USA
| | - Natalie Schmidt
- Physicians Committee for Responsible Medicine, Washington, DC, USA; Florida Atlantic University, Charles E. Schmidt College of Medicine, FL, USA
| | - Ellen Hata
- Physicians Committee for Responsible Medicine, Washington, DC, USA; George Washington University, Milken Institute School of Public Health, DC, USA
| | | | | | - Neal D Barnard
- Physicians Committee for Responsible Medicine, Washington, DC, USA; Adjunct faculty, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| |
Collapse
|
24
|
Woolf EK, Lee SY, Ghanem N, Vazquez AR, Johnson SA. Protective effects of blueberries on vascular function: A narrative review of preclinical and clinical evidence. Nutr Res 2023; 120:20-57. [PMID: 37913730 PMCID: PMC12046616 DOI: 10.1016/j.nutres.2023.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/18/2023] [Accepted: 09/23/2023] [Indexed: 11/03/2023]
Abstract
Blueberries are rich in nutrients and (poly)phenols, popular with consumers, and a major agricultural crop with year-round availability supporting their use in food-based strategies to promote human health. Accumulating evidence indicates blueberry consumption has protective effects on cardiovascular health including vascular dysfunction (i.e., endothelial dysfunction and arterial stiffening). This narrative review synthesizes evidence on blueberries and vascular function and provides insight into underlying mechanisms with a focus on oxidative stress, inflammation, and gut microbiota. Evidence from animal studies supports beneficial impacts on vascular function. Human studies indicate acute and chronic blueberry consumption can improve endothelial function in healthy and at-risk populations and may modulate arterial stiffness, but that evidence is less certain. Results from cell, animal, and human studies suggest blueberry consumption improves vascular function through improving nitric oxide bioavailability, oxidative stress, and inflammation. Limited data in animals suggest the gut microbiome mediates beneficial effects of blueberries on vascular function; however, there is a paucity of studies evaluating the gut microbiome in humans. Translational evidence indicates anthocyanin metabolites mediate effects of blueberries on endothelial function, though this does not exclude potential synergistic and/or additive effects of other blueberry components. Further research is needed to establish the clinical efficacy of blueberries to improve vascular function in diverse human populations in a manner that provides mechanistic information. Translation of clinical research to the community/public should consider feasibility, social determinants of health, culture, community needs, assets, and desires, barriers, and drivers to consumption, among other factors to establish real-world impacts of blueberry consumption.
Collapse
Affiliation(s)
- Emily K Woolf
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Sylvia Y Lee
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Nancy Ghanem
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Allegra R Vazquez
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Sarah A Johnson
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
25
|
Laurans L, Mouttoulingam N, Chajadine M, Lavelle A, Diedisheim M, Bacquer E, Creusot L, Suffee N, Esposito B, Melhem NJ, Le Goff W, Haddad Y, Paul JL, Rainteau D, Tedgui A, Ait-Oufella H, Zitvogel L, Sokol H, Taleb S. An obesogenic diet increases atherosclerosis through promoting microbiota dysbiosis-induced gut lymphocyte trafficking into the periphery. Cell Rep 2023; 42:113350. [PMID: 37897726 DOI: 10.1016/j.celrep.2023.113350] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
Although high-fat diet (HFD)-induced gut microbiota dysbiosis is known to affect atherosclerosis, the underlying mechanisms remain to be fully explored. Here, we show that the progression of atherosclerosis depends on a gut microbiota shaped by an HFD but not a high-cholesterol (HC) diet and, more particularly, on low fiber (LF) intake. Mechanistically, gut lymphoid cells impacted by HFD- or LF-induced microbiota dysbiosis highly proliferate in mesenteric lymph nodes (MLNs) and migrate from MLNs to the periphery, which fuels T cell accumulation within atherosclerotic plaques. This is associated with the induction of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) within plaques and the presence of enterotropic lymphocytes expressing β7 integrin. MLN resection or lymphocyte deficiency abrogates the pro-atherogenic effects of a microbiota shaped by LF. Our study shows a pathological link between a diet-shaped microbiota, gut immune cells, and atherosclerosis, suggesting that a diet-modulated microbiome might be a suitable therapeutic target to prevent atherosclerosis.
Collapse
Affiliation(s)
- Ludivine Laurans
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nirmala Mouttoulingam
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Mouna Chajadine
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Aonghus Lavelle
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), 37540 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades (INEM), Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, 75015 Paris, France
| | - Emilie Bacquer
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laura Creusot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Nadine Suffee
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Bruno Esposito
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nada Joe Melhem
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Wilfried Le Goff
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Yacine Haddad
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France
| | - Jean-Louis Paul
- Université Paris-Sud, Equipe d'Accueil 4529, UFR de Pharmacie, Chatenay-Malabry, France and Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Dominique Rainteau
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Clinical Metabolomics Department, 75012 Paris, France
| | - Alain Tedgui
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France; Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAe, Micalis & AgroParisTech, Jouy en Josas, France
| | - Soraya Taleb
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France.
| |
Collapse
|
26
|
Chen M, Peng L, Zhang C, Liu Q, Long T, Xie Q. Gut microbiota might mediate the benefits of high-fiber/acetate diet to cardiac hypertrophy mice. J Physiol Biochem 2023; 79:745-756. [PMID: 37537429 DOI: 10.1007/s13105-023-00971-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/28/2023] [Indexed: 08/05/2023]
Abstract
Continuously prolonged cardiac hypertrophy results in maladaptive myocardial remodeling, which affects cardiac function and can eventually lead to heart failure. Short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, have been reported to be associated with cardiovascular diseases (CVD). Gut microbiota may mediate between dietary fiber and SCFA effects on cardiac hypertrophy. The mice model of isoproterenol (ISO)-induced cardiac hypertrophy was constructed and verified for physiological, functional, and fibrotic alterations in this study. Both high-fiber and acetate diet improved physiological indexes, ameliorated cardiac functions, and relieved fibrotic alterations in model mice hearts; collectively, cardiac hypertrophy in mice receiving both high-fiber and acetate diet improved. Following 16s rDNA sequencing and integrative bioinformatics, analyses indicated that both high-fiber and acetate diet caused alterations in mice gut microbiota compared with the ISO group, including OTU composition and abundance. In conclusion, high-fiber and acetate diet improve the physiological status, cardiac functions, and fibrotic alterations in ISO-induced hypertrophic mice. Besides, considering the alterations in mice gut microbiota in response to single ISO, both high-fiber and acetate diet treatment, gut microbiota might mediate the favorable benefits of both high-fiber and acetate diet on cardiac hypertrophy.
Collapse
Affiliation(s)
- Meifang Chen
- Department of Geriatric Cardiology, Xiangya Hospital, Central South University, Changsha, 41008, China
- Department of Cardiology, Xiangya Hospital, Central South University, #87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
| | - Liming Peng
- Department of Cardiology, Xiangya Hospital, Central South University, #87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chenglong Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, #87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Liu
- Department of Cardiology, Xiangya Hospital, Central South University, #87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Tianyi Long
- Department of Cardiology, Xiangya Hospital, Central South University, #87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiying Xie
- Department of Cardiology, Xiangya Hospital, Central South University, #87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
27
|
Almeida C, Gonçalves-Nobre JG, Alpuim Costa D, Barata P. The potential links between human gut microbiota and cardiovascular health and disease - is there a gut-cardiovascular axis? FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1235126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut-heart axis is an emerging concept highlighting the crucial link between gut microbiota and cardiovascular diseases (CVDs). Recent studies have demonstrated that gut microbiota is pivotal in regulating host metabolism, inflammation, and immune function, critical drivers of CVD pathophysiology. Despite a strong link between gut microbiota and CVDs, this ecosystem’s complexity still needs to be fully understood. The short-chain fatty acids, trimethylamine N-oxide, bile acids, and polyamines are directly or indirectly involved in the development and prognosis of CVDs. This review explores the relationship between gut microbiota metabolites and CVDs, focusing on atherosclerosis and hypertension, and analyzes personalized microbiota-based modulation interventions, such as physical activity, diet, probiotics, prebiotics, and fecal microbiota transplantation, as a promising strategy for CVD prevention and treatment.
Collapse
|
28
|
Saxami G, Kerezoudi EN, Eliopoulos C, Arapoglou D, Kyriacou A. The Gut-Organ Axis within the Human Body: Gut Dysbiosis and the Role of Prebiotics. Life (Basel) 2023; 13:2023. [PMID: 37895405 PMCID: PMC10608660 DOI: 10.3390/life13102023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
The human gut microbiota (GM) is a complex microbial ecosystem that colonises the gastrointestinal tract (GIT) and is comprised of bacteria, viruses, fungi, and protozoa. The GM has a symbiotic relationship with its host that is fundamental for body homeostasis. The GM is not limited to the scope of the GIT, but there are bidirectional interactions between the GM and other organs, highlighting the concept of the "gut-organ axis". Any deviation from the normal composition of the GM, termed "microbial dysbiosis", is implicated in the pathogenesis of various diseases. Only a few studies have demonstrated a relationship between GM modifications and disease phenotypes, and it is still unknown whether an altered GM contributes to a disease or simply reflects its status. Restoration of the GM with probiotics and prebiotics has been postulated, but evidence for the effects of prebiotics is limited. Prebiotics are substrates that are "selectively utilized by host microorganisms, conferring a health benefit". This study highlights the bidirectional relationship between the gut and vital human organs and demonstrates the relationship between GM dysbiosis and the emergence of certain representative diseases. Finally, this article focuses on the potential of prebiotics as a target therapy to manipulate the GM and presents the gaps in the literature and research.
Collapse
Affiliation(s)
- Georgia Saxami
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.N.K.); (A.K.)
| | - Evangelia N. Kerezoudi
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.N.K.); (A.K.)
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden
| | - Christos Eliopoulos
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization—Demeter, L. Sof. Venizelou 1, 14123 Lykovryssi, Greece; (C.E.); (D.A.)
| | - Dimitrios Arapoglou
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization—Demeter, L. Sof. Venizelou 1, 14123 Lykovryssi, Greece; (C.E.); (D.A.)
| | - Adamantini Kyriacou
- Department of Nutrition and Dietetics, Harokopio University, 17671 Athens, Greece; (E.N.K.); (A.K.)
| |
Collapse
|
29
|
Dinu LD, Gatea F, Roaming Israel F, Lakicevic M, Dedović N, Vamanu E. The Modulation Effect of a Fermented Bee Pollen Postbiotic on Cardiovascular Microbiota and Therapeutic Perspectives. Biomedicines 2023; 11:2712. [PMID: 37893086 PMCID: PMC10604238 DOI: 10.3390/biomedicines11102712] [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: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Hypertension is a frequent comorbidity in patients with heart failure; therefore, blood pressure management for these patients is widely recommended in medical guidelines. Bee pollen and postbiotics that contain inactivated probiotic cells and their metabolites have emerged as promising bioactive compounds sources, and their potential role in mitigating cardiovascular (CV) risks is currently being unveiled. Therefore, this preliminary study aimed to investigate the impact of a lactic-fermented bee pollen postbiotic (FBPP) on the CV microbiota via in vitro tests. A new isolated Lactobacillus spp. strain from the digestive tract of bees was used to ferment pollen, obtaining liquid and dried atomized caps postbiotics. The modulating effects on a CV microbiota that corresponds to the pathophysiology of hypertension were investigated using microbiological methods and qPCR and correlated with the metabolic profile. Both liquid and dried FBPPs increased the number of the beneficial Lactobacillus spp. and Bifidobacterium spp. bacteria by up to 2 log/mL, while the opportunistic pathogen E. coli, which contributes to CV pathogenesis, decreased by 3 log/mL. The short-chain fatty acid (SCFA) profile revealed a significant increase in lactic (6.386 ± 0.106 g/L) and acetic (4.284 ± 0.017 g/L) acids, both with known antihypertensive effects, and the presence of isovaleric acid, which promotes a healthy gut microbiota. Understanding the impact of the FBPP on gut microbiota could lead to innovative strategies for promoting heart health and preventing cardiovascular diseases.
Collapse
Affiliation(s)
- Laura-Dorina Dinu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania;
| | - Florentina Gatea
- Centre of Bioanalysis, National Institute for Biological Sciences, 060031 Bucharest, Romania;
| | - Florentina Roaming Israel
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania;
| | - Milena Lakicevic
- Faculty of Agriculture, University of Novi Sad, 21000 Novi Sad, Serbia; (M.L.); (N.D.)
| | - Nebojša Dedović
- Faculty of Agriculture, University of Novi Sad, 21000 Novi Sad, Serbia; (M.L.); (N.D.)
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania;
| |
Collapse
|
30
|
Satheesh Babu AK, Srinivasan H, Anandh Babu PV. Breaking bugs: gut microbes metabolize dietary components and modulate vascular health. Crit Rev Food Sci Nutr 2023; 64:12411-12419. [PMID: 37651204 PMCID: PMC10902197 DOI: 10.1080/10408398.2023.2251616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Gut microbiota modulates host physiology and pathophysiology through the production of microbial metabolites. Diet is a crucial factor in shaping the microbiome, and gut microbes interact with the host by producing beneficial or detrimental diet-derived microbial metabolites. Evidence from our lab and others indicates that the interaction between diet and gut microbes plays a pivotal role in modulating vascular health. Diet-derived microbial metabolites such as short-chain fatty acids and metabolites of phenolic acids improve vascular health, whereas trimethylamine oxide and certain amino acid-derived microbial metabolites impair the vasculature. These metabolites have been shown to regulate blood pressure, vascular inflammation, and atherosclerosis by acting on multiple targets. Nonetheless, there are substantial gaps in knowledge within this field. The microbial enzymes essential for the production of diet-derived metabolites, the role of the food matrix in regulating the bioavailability of metabolites, and the structure-activity relationships between metabolites and biomolecules in the vasculature are largely unknown. Potential diet-derived metabolites to improve vascular health can be identified through future studies that investigate the causal relationship between dietary components, gut microbes, diet-derived metabolites, and vascular health by using radiolabeled compounds, metabolomics, transcriptomics, and proteomics techniques.
Collapse
Affiliation(s)
| | | | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
31
|
Nemet I, Li XS, Haghikia A, Li L, Wilcox J, Romano KA, Buffa JA, Witkowski M, Demuth I, König M, Steinhagen-Thiessen E, Bäckhed F, Fischbach MA, Tang WHW, Landmesser U, Hazen SL. Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality. Eur Heart J 2023; 44:3085-3096. [PMID: 37342006 PMCID: PMC10481777 DOI: 10.1093/eurheartj/ehad333] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
AIMS Precision microbiome modulation as a novel treatment strategy is a rapidly evolving and sought goal. The aim of this study is to determine relationships among systemic gut microbial metabolite levels and incident cardiovascular disease risks to identify gut microbial pathways as possible targets for personalized therapeutic interventions. METHODS AND RESULTS Stable isotope dilution mass spectrometry methods to quantitatively measure aromatic amino acids and their metabolites were used to examine sequential subjects undergoing elective diagnostic cardiac evaluation in two independent cohorts with longitudinal outcome data [US (n = 4000) and EU (n = 833) cohorts]. It was also used in plasma from humans and mice before vs. after a cocktail of poorly absorbed antibiotics to suppress gut microbiota. Multiple aromatic amino acid-derived metabolites that originate, at least in part, from gut bacteria are associated with incident (3-year) major adverse cardiovascular event (MACE) risks (myocardial infarction, stroke, or death) and all-cause mortality independent of traditional risk factors. Key gut microbiota-derived metabolites associated with incident MACE and poorer survival risks include: (i) phenylacetyl glutamine and phenylacetyl glycine (from phenylalanine); (ii) p-cresol (from tyrosine) yielding p-cresol sulfate and p-cresol glucuronide; (iii) 4-OH-phenyllactic acid (from tyrosine) yielding 4-OH-benzoic acid and 4-OH-hippuric acid; (iv) indole (from tryptophan) yielding indole glucuronide and indoxyl sulfate; (v) indole-3-pyruvic acid (from tryptophan) yielding indole-3-lactic acid and indole-3-acetyl-glutamine, and (vi) 5-OH-indole-3-acetic acid (from tryptophan). CONCLUSION Key gut microbiota-generated metabolites derived from aromatic amino acids independently associated with incident adverse cardiovascular outcomes are identified, and thus will help focus future studies on gut-microbial metabolic outputs relevant to host cardiovascular health.
Collapse
Affiliation(s)
- Ina Nemet
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Arash Haghikia
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin 12203, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin 10785, Germany
- Biomedical Innovation Academy, Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Lin Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jennifer Wilcox
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Kymberleigh A Romano
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jennifer A Buffa
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Marco Witkowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Center for Regenerative Therapies, Berlin Institute of Health (BIH), Berlin 13353, Germany
| | - Maximilian König
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | | | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg SE-413 45, Sweden
| | - Michael A Fischbach
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
- ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - W H Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin 12203, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin 10785, Germany
- Biomedical Innovation Academy, Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Center for Microbiome & Human Health, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
32
|
Yang Y, Karampoor S, Mirzaei R, Borozdkin L, Zhu P. The interplay between microbial metabolites and macrophages in cardiovascular diseases: A comprehensive review. Int Immunopharmacol 2023; 121:110546. [PMID: 37364331 DOI: 10.1016/j.intimp.2023.110546] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
The gut microbiome has emerged as a crucial player in developing and progressing cardiovascular diseases (CVDs). Recent studies have highlighted the role of microbial metabolites in modulating immune cell function and their impact on CVD. Macrophages, which have a significant function in the pathogenesis of CVD, are very vulnerable to the effects of microbial metabolites. Microbial metabolites, such as short-chain fatty acids (SCFAs) and trimethylamine-N-oxide (TMAO), have been linked to atherosclerosis and the regulation of immune functions. Butyrate has been demonstrated to reduce monocyte migration and inhibit monocyte attachment to injured endothelial cells, potentially contributing to the attenuation of the inflammatory response and the progression of atherosclerosis. On the other hand, TMAO, another compound generated by gut bacteria, has been linked to atherosclerosis due to its impact on lipid metabolism and the accumulation of cholesterol in macrophages. Indole-3-propionic acid, a tryptophan metabolite produced solely by microbes, has been found to promote the development of atherosclerosis by stimulating macrophage reverse cholesterol transport (RCT) and raising the expression of ABCA1. This review comprehensively discusses how various microbiota-produced metabolites affect macrophage polarization, inflammation, and foam cell formation in CVD. We also highlight the mechanisms underlying these effects and the potential therapeutic applications of targeting microbial metabolites in treating CVD.
Collapse
Affiliation(s)
- Yongzheng Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Leonid Borozdkin
- Department of Maxillofacial Surgery, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
33
|
Wang Q, Yesitayi G, Liu B, Siti D, Ainiwan M, Aizitiaili A, Ma X. Targeting metabolism in aortic aneurysm and dissection: from basic research to clinical applications. Int J Biol Sci 2023; 19:3869-3891. [PMID: 37564200 PMCID: PMC10411465 DOI: 10.7150/ijbs.85467] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
Aortic aneurysm and dissection (AAD) are a group of insidious and lethal cardiovascular diseases that characterized by seriously threatening the life and health of people, but lack effective nonsurgical interventions. Alterations in metabolites are increasingly recognized as universal features of AAD because metabolic abnormalities have been identified not only in arterial tissue but also in blood and vascular cells from both patients and animal models with this disease. Over the past few decades, studies have further supported this notion by linking AAD to various types of metabolites such as those derived from gut microbiota or involved in TCA cycle or lipid metabolism. Many of these altered metabolites may contribute to the pathogenesis of AAD. This review aims to illustrate the close association between body metabolism and the occurrence and development of AAD, as well as summarize the significance of metabolites correlated with the pathological process of AAD. This provides valuable insight for developing new therapeutic agents for AAD. Therefore, we present a brief overview of metabolism in AAD biology, including signaling pathways involved in these processes and current clinical studies targeting AAD metabolisms. It is necessary to understand the metabolic mechanisms underlying AAD to provides significant knowledge for AAD diagnosis and new therapeutics for treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Gulinazi Yesitayi
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Bingyan Liu
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Dilixiati Siti
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Mierxiati Ainiwan
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Aliya Aizitiaili
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| | - Xiang Ma
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
34
|
Endres K, Friedland K. Talk to Me-Interplay between Mitochondria and Microbiota in Aging. Int J Mol Sci 2023; 24:10818. [PMID: 37445995 DOI: 10.3390/ijms241310818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The existence of mitochondria in eukaryotic host cells as a remnant of former microbial organisms has been widely accepted, as has their fundamental role in several diseases and physiological aging. In recent years, it has become clear that the health, aging, and life span of multicellular hosts are also highly dependent on the still-residing microbiota, e.g., those within the intestinal system. Due to the common evolutionary origin of mitochondria and these microbial commensals, it is intriguing to investigate if there might be a crosstalk based on preserved common properties. In the light of rising knowledge on the gut-brain axis, such crosstalk might severely affect brain homeostasis in aging, as neuronal tissue has a high energy demand and low tolerance for according functional decline. In this review, we summarize what is known about the impact of both mitochondria and the microbiome on the host's aging process and what is known about the aging of both entities. For a long time, bacteria were assumed to be immortal; however, recent evidence indicates their aging and similar observations have been made for mitochondria. Finally, we present pathways by which mitochondria are affected by microbiota and give information about therapeutic anti-aging approaches that are based on current knowledge.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Kristina Friedland
- Department of Pharmacology and Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University, 55128 Mainz, Germany
| |
Collapse
|
35
|
Zhen J, Zhou Z, He M, Han HX, Lv EH, Wen PB, Liu X, Wang YT, Cai XC, Tian JQ, Zhang MY, Xiao L, Kang XX. The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne) 2023; 14:1085041. [PMID: 36824355 PMCID: PMC9941174 DOI: 10.3389/fendo.2023.1085041] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Morbidity and mortality of cardiovascular diseases (CVDs) are exceedingly high worldwide. Researchers have found that the occurrence and development of CVDs are closely related to intestinal microecology. Imbalances in intestinal microecology caused by changes in the composition of the intestinal microbiota will eventually alter intestinal metabolites, thus transforming the host physiological state from healthy mode to pathological mode. Trimethylamine N-oxide (TMAO) is produced from the metabolism of dietary choline and L-carnitine by intestinal microbiota, and many studies have shown that this important product inhibits cholesterol metabolism, induces platelet aggregation and thrombosis, and promotes atherosclerosis. TMAO is directly or indirectly involved in the pathogenesis of CVDs and is an important risk factor affecting the occurrence and even prognosis of CVDs. This review presents the biological and chemical characteristics of TMAO, and the process of TMAO produced by gut microbiota. In particular, the review focuses on summarizing how the increase of gut microbial metabolite TMAO affects CVDs including atherosclerosis, heart failure, hypertension, arrhythmia, coronary artery disease, and other CVD-related diseases. Understanding the mechanism of how increases in TMAO promotes CVDs will potentially facilitate the identification and development of targeted therapy for CVDs.
Collapse
Affiliation(s)
- Jing Zhen
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Chemical Engineering and Technology, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Zhou Zhou
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng He
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hai-Xiang Han
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - En-Hui Lv
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng-Bo Wen
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Liu
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan-Ting Wang
- Department of Biochemical Pharmacy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Xun-Chao Cai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
| | - Jia-Qi Tian
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng-Ying Zhang
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Xiao
- School of Chemical Engineering and Technology, China University of Mining and Technology, Xuzhou, Jiangsu, China
| | - Xing-Xing Kang
- Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
36
|
Rinninella E, Tohumcu E, Raoul P, Fiorani M, Cintoni M, Mele MC, Cammarota G, Gasbarrini A, Ianiro G. The role of diet in shaping human gut microbiota. Best Pract Res Clin Gastroenterol 2023; 62-63:101828. [PMID: 37094913 DOI: 10.1016/j.bpg.2023.101828] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/26/2023]
Abstract
Gut microbiota plays a fundamental role within human health, and exerts key functions within the human body. Diet is one of the most powerful modulators of gut microbiota functions and composition. This complex interplay involves also the immune system and the intestinal barrier, highlighting the central role of diet in the pathogenesis and treatment of multiple diseases. In this review article we will paint the landscape of the effects of specific dietary nutrients, and of the detrimental or beneficial outcomes of different dietary patterns, on the composition of human gut microbiota. Moreover, we will discuss the potential application of diet as a therapeutic modulator of gut microbiota, including cutting-edge ways of exploitation, including the use of dietary components as adjuvants to promote microbial engraftment after fecal microbiota transplantation, or personalized nutritional approaches, targeted to the patient microbiome.
Collapse
Affiliation(s)
- Emanuele Rinninella
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ege Tohumcu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Pauline Raoul
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marcello Fiorani
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marco Cintoni
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Cristina Mele
- Department of Medical and Surgical Sciences, Clinical Nutrition Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
37
|
Inflammatory Response: A Crucial Way for Gut Microbes to Regulate Cardiovascular Diseases. Nutrients 2023; 15:nu15030607. [PMID: 36771313 PMCID: PMC9921390 DOI: 10.3390/nu15030607] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/09/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Gut microbiota is the largest and most complex microflora in the human body, which plays a crucial role in human health and disease. Over the past 20 years, the bidirectional communication between gut microbiota and extra-intestinal organs has been extensively studied. A better comprehension of the alternative mechanisms for physiological and pathophysiological processes could pave the way for health. Cardiovascular disease (CVD) is one of the most common diseases that seriously threatens human health. Although previous studies have shown that cardiovascular diseases, such as heart failure, hypertension, and coronary atherosclerosis, are closely related to gut microbiota, limited understanding of the complex pathogenesis leads to poor effectiveness of clinical treatment. Dysregulation of inflammation always accounts for the damaged gastrointestinal function and deranged interaction with the cardiovascular system. This review focuses on the characteristics of gut microbiota in CVD and the significance of inflammation regulation during the whole process. In addition, strategies to prevent and treat CVD through proper regulation of gut microbiota and its metabolites are also discussed.
Collapse
|
38
|
Dietary Methionine Restriction Alleviates Choline-Induced Tri-Methylamine-N-Oxide (TMAO) Elevation by Manipulating Gut Microbiota in Mice. Nutrients 2023; 15:nu15010206. [PMID: 36615863 PMCID: PMC9823801 DOI: 10.3390/nu15010206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/14/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Dietary methionine restriction (MR) has been shown to decrease plasma trimethylamine-N-oxide (TMAO) levels in high-fat diet mice; however, the specific mechanism used is unknown. We speculated that the underlying mechanism is related with the gut microbiota, and this study aimed to confirm the hypothesis. In this study, we initially carried out an in vitro fermentation experiment and found that MR could reduce the ability of gut microbiota found in the contents of healthy mice and the feces of healthy humans to produce trimethylamine (TMA). Subsequently, mice were fed a normal diet (CON, 0.20% choline + 0.86% methionine), high-choline diet (H-CHO, 1.20% choline + 0.86% methionine), or high-choline + methionine-restricted diet (H-CHO+MR, 1.20% choline + 0.17% methionine) for 3 months. Our results revealed that MR decreased plasma TMA and TMAO levels in H-CHO-diet-fed mice without changing hepatic FMO3 gene expression and enzyme activity, significantly decreased TMA levels and expression of choline TMA-lyase (CutC) and its activator CutD, and decreased CutC activity in the intestine. Moreover, MR significantly decreased the abundance of TMA-producing bacteria, including Escherichia-Shigella (Proteobacteria phylum) and Anaerococcus (Firmicutes phylum), and significantly increased the abundance of short-chain fatty acid (SCFA)-producing bacteria and SCFA levels. Furthermore, both MR and sodium butyrate supplementation significantly inhibited bacterial growth, down-regulated CutC gene expression levels in TMA-producing bacteria, including Escherichia fergusonii ATCC 35469 and Anaerococcus hydrogenalis DSM 7454 and decreased TMA production from bacterial growth under in vitro anaerobic fermentation conditions. In conclusion, dietary MR alleviates choline-induced TMAO elevation by manipulating gut microbiota in mice and may be a promising approach to reducing circulating TMAO levels and TMAO-induced atherosclerosis.
Collapse
|
39
|
Threonine Facilitates Cd Excretion by Increasing the Abundance of Gut Escherichia coli in Cd-Exposed Mice. Molecules 2022; 28:molecules28010177. [PMID: 36615370 PMCID: PMC9822384 DOI: 10.3390/molecules28010177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Cadmium (Cd) can easily enter the body through the food chain and threaten health since Cd pollution is prevalent in the environment. Gut microbiota is necessary for the reduction of metal ions. To reduce Cd-induced harmful impacts and Cd accumulation in the body, we investigated the effect of amino acids on gut microbiota and Cd excretion in (fecal Cd) Cd-exposed mice. The screening of 20 amino acids showed that threonine (Thr) effectively increased fecal Cd, and reduced Cd-induced intestinal structural damage. The abundance of Escherichia-Shigella genus and KF843036_g significantly increased after the oral administration of Thr. As the type species of the Escherichia-Shigella genus, Escherichia coli exhibited high similarity to KF843036_g species and significantly decreased Cd-induced gut damage. Cd contents in the liver, kidney, and gut of Cd-exposed mice were also significantly (p < 0.05) decreased after E. coli treatment, while the contents in the feces were increased. The results demonstrated the potential roles that gut E. coli might play in Thr-mediated Cd excretion in Cd-exposed mice. The findings may provide important data for better understanding the molecular biological mechanism of Thr in reducing Cd accumulation in the body.
Collapse
|
40
|
Yaskolka Meir A, Liang L. The gut microbiota's role in diet-related cardiovascular health: an innocent bystander or a key mediator; the question remains. Eur J Prev Cardiol 2022; 29:1893-1894. [PMID: 35929653 DOI: 10.1093/eurjpc/zwac167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Anat Yaskolka Meir
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
41
|
Kato-Kogoe N, Kamiya K, Sakaguchi S, Omori M, Komori E, Kudo A, Nakamura S, Nakano T, Ueno T, Tamaki J, Hoshiga M. Salivary Microbiota Associated with Peripheral Microvascular Endothelial Dysfunction. J Atheroscler Thromb 2022. [PMID: 36130883 DOI: 10.5551/jat.63681] [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: 11/11/2022] Open
Abstract
AIMS Oral health is associated with atherosclerotic cardiovascular disease (ACVD). We previously identified the salivary microbiota characteristics of patients with ACVD. However, whether salivary microbiota is characteristic under impaired vascular endothelial function before ACVD onset remains unclear. Therefore, we aimed to evaluate the characteristics of salivary microbiota associated with peripheral microvascular endothelial dysfunction. METHODS We collected saliva samples from 172 community-dwelling elderly individuals without a history of ACVD and performed 16S rRNA metagenomic analysis. We assessed the peripheral microvascular endothelial function using reactive hyperemia index (RHI) and compared the salivary microbiota in the groups with normal (RHI ≥ 2.10), borderline, and abnormal (RHI <1.67) peripheral endothelial function. Furthermore, we applied machine learning techniques to evaluate whether salivary microbiota could discriminate between individuals with normal and abnormal endothelial function. RESULTS The number of operational taxonomic units (OTUs) was higher in the abnormal group than in the normal group (p=0.037), and differences were found in the overall salivary microbiota structure (unweighted UniFrac distances, p=0.038). The linear discriminant analysis (LDA) effect size (LEfSe) algorithm revealed several significantly differentially abundant bacterial genera between the two groups. An Extra Trees classifier model was built to discriminate between groups with normal and abnormal vascular endothelial function based on the microbial composition at the genus level (AUC=0.810). CONCLUSIONS The salivary microbiota in individuals with endothelial dysfunction was distinct from that in individuals with normal endothelial function, indicating that the salivary microbiota may be related to endothelial function.
Collapse
Affiliation(s)
- Nahoko Kato-Kogoe
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Kuniyasu Kamiya
- Department of Hygiene and Public Health, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Shoichi Sakaguchi
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Michi Omori
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Eri Komori
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Asako Kudo
- Department of Hygiene and Public Health, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University
| | - Takashi Nakano
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Takaaki Ueno
- Department of Dentistry and Oral Surgery, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Junko Tamaki
- Department of Hygiene and Public Health, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| | - Masaoki Hoshiga
- Department of Cardiology, Faculty of Medicine, Osaka Medical and Pharmaceutical University
| |
Collapse
|
42
|
Mahoney DE, Chalise P, Rahman F, Pierce JD. Influences of Gastrointestinal Microbiota Dysbiosis on Serum Proinflammatory Markers in Epithelial Ovarian Cancer Development and Progression. Cancers (Basel) 2022; 14:3022. [PMID: 35740687 PMCID: PMC9220985 DOI: 10.3390/cancers14123022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 01/27/2023] Open
Abstract
GI microbiota has been implicated in producing the inflammatory tumor microenvironment of several cancers. Women with ovarian cancer often report GI-related symptoms at diagnosis although minimal is known about the possible GI bacteria that may trigger pro-tumorigenic immune responses in early EOC. The purpose of this study was to investigate the influences of GI microbiota dysbiosis on serum inflammatory markers during EOC utilizing a rodent model. This experimental design consisted of C57BL/6 mice randomly assigned to either the microbiota dysbiosis group (n = 6) or control group (n = 5). The CD7BL/6 mice assigned to the microbiota dysbiosis group were administered a mixture of broad-spectrum antibiotics (bacitracin and neomycin) for 2 weeks. Both groups were injected intraperitoneally with mouse ovarian epithelial cells that induce ovarian tumorigenesis. Levels of C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) were assessed in the serum, and the composition of the GI microbiota in fecal samples was measured using 16S rRNA gene sequencing. Overall CRP serum levels were significantly lower and TNFα levels were significantly higher in the microbiota dysbiosis group compared to the control group. The abundances of microbiota that correlated with CRP serum levels in the combined groups were genus Parabacteroides, Roseburia, and Emergencia and species Ruminococcus faecis, Parabacteroides distasonis, Roseburia Faecis, and Emergencia timonensis. This study provides evidence to support for further investigation of the GI microbial profiles in patients at risk of EOC.
Collapse
Affiliation(s)
- Diane E. Mahoney
- School of Nursing, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Prabhakar Chalise
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Faith Rahman
- Clinical Trials Clinical Operations, University of Kansas Cancer Center, Kansas City, KS 66160, USA;
| | - Janet D. Pierce
- School of Nursing, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| |
Collapse
|