1
|
Tang L, Li J, Luan M, Qin M, Zhong C, Zhang Y, Xie Y, Shi M, Qiu L, Yu J. Edgeworthia gardneri (Wall.) Meisn protects against HFD-induced murine atherosclerosis through improving gut microbiota-mediated intestinal barrier integrity. Atherosclerosis 2025; 403:119132. [PMID: 40015156 DOI: 10.1016/j.atherosclerosis.2025.119132] [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: 09/12/2024] [Revised: 01/18/2025] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Gut microbiota plays a crucial role in the development and progression of atherosclerosis. Edgeworthia gardneri (Wall.) Meisn, a member of the Thymelaeaceae family and the Edgeworthia genus, has been previously shown in our studies to attenuate atherogenesis when administered orally as an ethanolic extract (EEEG). However, the interaction between EEEG and gut microbiota, and the mechanism by which gut microbiota exerts anti-atherosclerotic effects, remains unclear. AIMS This study aims to determine whether the anti-atherosclerotic properties of EEEG are associated with gut microbiota remodeling. METHOD Atherosclerosis was induced in ApoE-/- mice using a high-fat diet (HFD). The mice were treated with EEEG or Lactobacillus plantarum for 16 weeks. The composition of gut microbiota was analyzed through 16S rDNA sequencing. To assess whether the anti-atherosclerotic effects of EEEG depend on the gut microbiota, HFD-fed mice were treated with a cocktail of antibiotics or underwent fecal microbiota transplantation (FMT). Simultaneously, plaque areas in the aortic roots and whole aortas of apolipoprotein E deficient (ApoE-/-) mice were evaluated using oil red O staining and hematoxylin-eosin staining. Serum levels of LPS, fluorescein isothiocyanate-dextran, and expression levels of tight junction proteins were measured to identify the effects of EEEG on gut barrier dysfunction in HFD-fed ApoE-/- mice. RESULTS The results revealed that EEEG treatment significantly reduced atherosclerotic lesions by ameliorating lipid accumulation and preserving gut barrier integrity. The protective effects were abrogated by antibiotics administration, concomitant with an increase in gut barrier permeability by decreasing expression of tight junction proteins. The microbial analysis indicated an augmented abundance of Lactobacillus, Turicibacter, Faecalibacterium, Akkermansia, and Desulfovibrio following EEEG treatment. Meanwhile, transplantation of fecal microbiota from EEEG-treated mice exerted the anti-atherosclerotic effect in the high-fat diet (HFD)-fed ApoE-/- recipient mice, accompanied by improvement of gut barrier integrity through upregulation of tight junction protein expression. Furthermore, exogenous supplementation of Lactobacillus plantarum mitigated AS in ApoE-/- mice and improved the gut epithelial barrier function by increasing the expression level of Zo-1. CONCLUSION These results suggest that the anti-atherosclerotic efficacy of EEEG is attributed to the preservation of gut barrier integrity mediated by gut microbiota. EEEG and its enriched Lactobacillus plantarum may be promising adjuncts for AS management. IMPORTANCE Atherosclerosis (AS) is the primary pathological basis of cardiovascular disease (CVD). The gut microbiota is known to play an important role in the development and progression of atherosclerosis. In the clinical management of AS, pharmacological classes such as antioxidants, lipid-lowering drugs, and antiplatelet agents are commonly utilized. Despite their ability to decelerate the progression of AS, complications and adverse reactions still limit their application. Edgeworthia gardneri (Wall.) Meisn, a member of the Thymelaeaceae family and Edgeworthia Meisn genus, has been shown in previous studies to attenuate atherogenesis when orally administered as an ethanolic extract (EEEG). However, the interaction between EEEG and the gut microbiota, as well as the mechanism by which the gut microbiota exerts its anti-atherosclerotic effects, remain unclear. The significance of our research lies in identifying the mechanism behind the anti-atherosclerotic effect of Edgeworthia gardneri. The expected results will provide an important scientific basis for the clinical development and application of Edgeworthia gardneri in the prevention and treatment of AS.
Collapse
Affiliation(s)
- Le Tang
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Jiangsheng Li
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Mingxuan Luan
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Manman Qin
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Chao Zhong
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Yifeng Zhang
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Yanfei Xie
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Min Shi
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Liang Qiu
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodeling Diseases, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Jun Yu
- Department of Cardiovascular Sciences and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
2
|
Wu W, Sun Y, Niu S, Li X, Chen L, Xie S, Chang L, Wei S, Jing M, Li H, Zhao Y. Integrated Microbiome and Metabolomic to Explore the Mechanism of Coptisine in Alleviating Ulcerative Colitis. Phytother Res 2025; 39:676-697. [PMID: 39648789 PMCID: PMC11832363 DOI: 10.1002/ptr.8389] [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: 12/12/2023] [Revised: 08/20/2024] [Accepted: 11/03/2024] [Indexed: 12/10/2024]
Abstract
Coptisine (COP), a naturally occurring alkaloid, is known for its diverse pharmacological effects and its supportive role in intestinal health. Despite this, the detailed mechanisms behind its therapeutic benefits are not yet fully understood. The objective of this study is to investigate the therapeutic potential of COP for the treatment of Ulcerative Colitis (UC) and to delineate the critical pathways by which it exerts its therapeutic effects. To assess COP's therapeutic effectiveness, mice were administered COP and monitored for clinical symptoms, activity, and disease activity index (DAI) changes. Intestinal histopathology, mucosal barrier function, and gut microbiota structure were evaluated, along with metabolic profiling, focusing on Prenol lipids in the colon to identify COP-induced metabolic shifts. Mice treated with COP exhibited significant relief from diarrhea and bleeding, along with increased activity and a marked reduction in DAI scores. Histopathological evaluation revealed a reduction in intestinal inflammation, and the intestinal mucosal barrier function was notably enhanced. The gut microbiota composition in COP-treated mice showed improvements. Additionally, the levels of Prenol lipids in the colon were elevated by COP treatment, which is crucial for the recovery of intestinal function. Our study demonstrates that COP effectively ameliorates colitis symptoms by modulating colon Prenol lipids metabolism, particularly under the influence of key bacterial species. The findings of this study provide novel insights into the therapeutic mechanisms of COP in the treatment of UC.
Collapse
Affiliation(s)
- Wenbin Wu
- Graduate School of Chinese PLA General HospitalChinese PLA Medical SchoolBeijingChina
- Health Care Office of the Service Bureau of AgencyOffices Administration of the Central Military CommissionBeijingChina
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Yanling Sun
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Shengqi Niu
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Xing Li
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Lisheng Chen
- College of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Shuying Xie
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Lei Chang
- School of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouChina
| | - Shizhang Wei
- National Cancer CenterNational Clinical Research Center for CancerCancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Manyi Jing
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Haotian Li
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| | - Yanling Zhao
- The Fifth Medical CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
3
|
Nunzi E, Pariano M, Costantini C, Garaci E, Puccetti P, Romani L. Host-microbe serotonin metabolism. Trends Endocrinol Metab 2025; 36:83-95. [PMID: 39142913 DOI: 10.1016/j.tem.2024.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024]
Abstract
As a result of a long evolutionary history, serotonin plays a variety of physiological roles, including neurological, cardiovascular, gastrointestinal, and endocrine functions. While many of these activities can be accommodated within the serotoninergic activity, recent findings have revealed an unsuspected role of serotonin in orchestrating host and microbial dialogue at the tryptophan dining table, to the benefit of local and systemic homeostasis. Herein we review the dual role of serotonin at the host-microbe interface and discuss how unraveling the interconnections among the host and microbial pathways of tryptophan degradation may help to accommodate the versatility of serotonin in physiology and pathology.
Collapse
Affiliation(s)
- Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; Casa di cura San Raffaele, Sulmona, L'Aquila, Italy.
| |
Collapse
|
4
|
Shayista H, Prasad MN, Raj SN, Ranjini H, Manju K, Baker S. Mechanistic overview of gut microbiota and mucosal pathogens with respect to cardiovascular diseases. THE MICROBE 2024; 5:100160. [DOI: 10.1016/j.microb.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Guarner F, Bustos Fernandez L, Cruchet S, Damião A, Maruy Saito A, Riveros Lopez JP, Rodrigues Silva L, Valdovinos Diaz MA. Gut dysbiosis mediates the association between antibiotic exposure and chronic disease. Front Med (Lausanne) 2024; 11:1477882. [PMID: 39568738 PMCID: PMC11576192 DOI: 10.3389/fmed.2024.1477882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Antibiotics are safe, effective drugs and continue to save millions of lives and prevent long-term illness worldwide. A large body of epidemiological, interventional and experimental evidence shows that exposure to antibiotics has long-term negative effects on human health. We reviewed the literature data on the links between antibiotic exposure, gut dysbiosis, and chronic disease (notably with regard to the "developmental origins of health and disease" ("DOHaD") approach). Molecular biology studies show that the systemic administration of antibiotic to infants has a rapid onset but also often a long-lasting impact on the microbial composition of the gut. Along with other environmental factors (e.g., an unhealthy "Western" diet and sedentary behavior), antibiotics induce gut dysbiosis, which can be defined as the disruption of a previously stable, functionally complete microbiota. Gut dysbiosis many harmful long-term effects on health. Associations between early-life exposure to antibiotics have been reported for chronic diseases, including inflammatory bowel disease, celiac disease, some cancers, metabolic diseases (obesity and type 2 diabetes), allergic diseases, autoimmune disorders, atherosclerosis, arthritis, and neurodevelopmental, neurodegenerative and other neurological diseases. In mechanistic terms, gut dysbiosis influences chronic disease through direct effects on mucosal immune and inflammatory pathways, plus a wide array of direct or indirect effects of short-chain fatty acids, the enteric nervous system, peristaltic motility, the production of hormones and neurotransmitters, and the loss of intestinal barrier integrity (notably with leakage of the pro-inflammatory endotoxin lipopolysaccharide into the circulation). To mitigate dysbiosis, the administration of probiotics in patients with chronic disease is often (but not always) associated with positive effects on clinical markers (e.g., disease scores) and biomarkers of inflammation and immune activation. Meta-analyses are complicated by differences in probiotic composition, dose level, and treatment duration, and large, randomized, controlled clinical trials are lacking in many disease areas. In view of the critical importance of deciding whether or not to prescribe antibiotics (especially to children), we suggest that the DOHaD concept can be logically extended to "gastrointestinal origins of health and disease" ("GOHaD") or even "microbiotic origins of health and disease" ("MOHaD").
Collapse
Affiliation(s)
| | - Luis Bustos Fernandez
- Centro Medico Bustos Fernandez, Instituto de Gastroenterologia, Buenos Aires, Argentina
| | - Sylvia Cruchet
- Institute of Nutrition and Food Technology, Universidad de Chile, Santiago, Chile
| | - Adérson Damião
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aldo Maruy Saito
- Catedra de Pediatria, Hospital Cayetano Heredia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | |
Collapse
|
6
|
Sha Y, Liu X, Li X, Wang Z, Shao P, Jiao T, He Y, Zhao S. Succession of rumen microbiota and metabolites across different reproductive periods in different sheep breeds and their impact on the growth and development of offspring lambs. MICROBIOME 2024; 12:172. [PMID: 39267132 PMCID: PMC11397069 DOI: 10.1186/s40168-024-01892-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/30/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND The microbiota and metabolites in the gastrointestinal tracts of female animals at different reproductive periods are very important to the growth, development, and health of themselves and their offspring. However, the changes in the gastrointestinal microbiota and metabolites throughout reproductive period of different sheep breeds and their effects on the growth and development of offspring lambs are still unclear. Hence, this study presents an assessment of the reproductive hormone levels, immune levels, rumen microbiota, and metabolites in Hu sheep and Suffolk ewes at different reproductive periods and their effects on the growth and development of offspring lambs. RESULTS Hu sheep and Suffolk during non-pregnancy, pregnancy, and lactation were used as the research objects to determine reproductive and immune indexes of ewes at different periods, analyze rumen microbiome and metabolome, and track the growth performance and development of offspring lambs. The results showed that the reproductive hormone and immune levels of Hu sheep and Suffolk underwent adaptive changes across different reproductive periods. Compared with non-pregnancy, the microbial energy metabolism and lipid metabolism function decreased during Hu sheep pregnancy, and energy metabolism function decreased during lactation. In Suffolk, energy metabolism, glycan biosynthesis, and metabolism function were enhanced during pregnancy, and the metabolism of cofactors and vitamins was enhanced during lactation. Prevotella increased in Suffolk during pregnancy and lactation (P < 0.05) and was positively correlated with the birth weight and body size of the lambs (P < 0.05). Moreover, the abundances of Butyrivibrio and Rikenellaceae_RC9_gut_group during pregnancy were positively correlated with the intestinal immunity of the offspring lambs (P < 0.05), thereby regulating the intestinal immunity level of the lambs. Metabolomic analysis revealed that the protein digestion, absorption, and amino acid metabolism of Hu sheep were enhanced during pregnancy, which provided amino acids for the growth and development of pregnant ewes and fetuses and was significantly correlated with the birth weight, body size, and intestinal immunity of lambs (P < 0.05). Simultaneously, there was an increase in acetate and propionate during the pregnancy and lactation period of both Hu sheep and Suffolk, providing energy for ewes during reproductive period. Moreover, the microbiota during the lactation period was significantly correlated with the milk quality and lambs daily gain (P < 0.05). CONCLUSIONS This study revealed the characteristic succession changes in the rumen microbiota and its metabolites at different reproductive periods in sheep breeds and their regulation of reproductive hormone and immune levels and identified their potential effects on the growth and development of offspring lambs. The findings provide valuable insights into the health and feeding management of different sheep breeds during the reproductive stage. Video Abstract.
Collapse
Affiliation(s)
- Yuzhu Sha
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiu Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiongxiong Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhengwen Wang
- College of Pratacultural Science, Gansu Agricultural University / Key Laboratory for Grassland Ecosystem, Ministry of Education / Sino-US Grassland Animal Husbandry Sustainable Development Research Center, Lanzhou, 730070, China
| | - Pengyang Shao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Ting Jiao
- College of Pratacultural Science, Gansu Agricultural University / Key Laboratory for Grassland Ecosystem, Ministry of Education / Sino-US Grassland Animal Husbandry Sustainable Development Research Center, Lanzhou, 730070, China
| | - Yanyu He
- School of Fundamental Sciences, Massey University, Palmerston North, 4410, New Zealand
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
7
|
Pi Y, Fang M, Li Y, Cai L, Han R, Sun W, Jiang X, Chen L, Du J, Zhu Z, Li X. Interactions between Gut Microbiota and Natural Bioactive Polysaccharides in Metabolic Diseases: Review. Nutrients 2024; 16:2838. [PMID: 39275156 PMCID: PMC11397228 DOI: 10.3390/nu16172838] [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: 07/16/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
The gut microbiota constitutes a complex ecosystem, comprising trillions of microbes that have co-evolved with their host over hundreds of millions of years. Over the past decade, a growing body of knowledge has underscored the intricate connections among diet, gut microbiota, and human health. Bioactive polysaccharides (BPs) from natural sources like medicinal plants, seaweeds, and fungi have diverse biological functions including antioxidant, immunoregulatory, and metabolic activities. Their effects are closely tied to the gut microbiota, which metabolizes BPs into health-influencing compounds. Understanding how BPs and gut microbiota interact is critical for harnessing their potential health benefits. This review provides an overview of the human gut microbiota, focusing on its role in metabolic diseases like obesity, type II diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular diseases. It explores the basic characteristics of several BPs and their impact on gut microbiota. Given their significance for human health, we summarize the biological functions of these BPs, particularly in terms of immunoregulatory activities, blood sugar, and hypolipidemic effect, thus providing a valuable reference for understanding the potential benefits of natural BPs in treating metabolic diseases. These properties make BPs promising agents for preventing and treating metabolic diseases. The comprehensive understanding of the mechanisms by which BPs exert their effects through gut microbiota opens new avenues for developing targeted therapies to improve metabolic health.
Collapse
Affiliation(s)
- Yu Pi
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Miaoyu Fang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Yanpin Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Long Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Ruyi Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenjuan Sun
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xianren Jiang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Zhigang Zhu
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
8
|
Liu Y, Wang Y, Wang H. Effects of ciprofloxacin and levofloxacin on initial colonization of intestinal microbiota in Bufo gargarizans at embryonic stages. CHEMOSPHERE 2024; 361:142587. [PMID: 38871193 DOI: 10.1016/j.chemosphere.2024.142587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Ciprofloxacin (CIP) and levofloxacin (LEV) are broad-spectrum antibiotics with potent antibacterial activity. Although many studies have shown that antibiotics can lead to gut microbiota disruption, the effects of CIP and LEV on gut microbial colonization at the embryonic stage remain poorly characterized. Here, we evaluated the response of Bufo gargarizans embryos in terms of gut microbiota colonization, growth and developmental stages to CIP and LEV exposure. Embryos treated with 100 μg/L CIP and LEV exhibited significantly reduced diversity and richness of the gut microbiota, as well as altered community structure. Both CIP and LEV treatments resulted in an increase in the pathogenic bacteria Bosea and Aeromonas, and they appeared to be more resistant to CIP than LEV. Additionally, CIP exposure caused reduced total length and delayed the development in B. gargarizans embryos, while LEV increased the total length and promoted embryonic development. The present study revealed the adverse effects of CIP and LEV exposure on host gut microbiota, growth and development during the embryonic stage, and contributed new perspectives to the evaluation of early aquatic ecological risk under CIP and LEV exposure.
Collapse
Affiliation(s)
- Ying Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| | - Yaxi Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| |
Collapse
|
9
|
Wang Z, Guo Z, Liu L, Ren D, Zu H, Li B, Liu F. Potential Probiotic Weizmannia coagulans WC10 Improved Antibiotic-Associated Diarrhea in Mice by Regulating the Gut Microbiota and Metabolic Homeostasis. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10308-1. [PMID: 38900235 DOI: 10.1007/s12602-024-10308-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Antibiotic-associated diarrhea (AAD) is a common side effect of long-term and heavy antibiotic therapy. Weizmannia coagulans (W. coagulans) is an ideal probiotic because of its high viability, stability, and numerous health benefits to the host. In this study, the strains were first screened for W. coagulans WC10 (WC10) with a high combined ability based on their biological properties of gastrointestinal tolerance, adhesion, and short-chain fatty acid production ability. The effect of WC10 on mice with AAD was further evaluated. The results showed that WC10 was effective in improving the symptoms of AAD, effectively restoring antibiotic-induced weight loss, and reducing diarrhea status score and fecal water content. In addition, WC10 decreased the expression of pro-inflammatory cytokines and increased the expression of anti-inflammatory cytokines, alleviated intestinal tissue damage and inflammation, and improved intestinal epithelial barrier function by decreasing serum levels of enterotoxin, DAO, and D-lactic acid, and by increasing the expression of the intestinal mucosal immune factors sIgA and occludin. Importantly, the composition and function of the gut microbiota gradually recovered after WC10 treatment, increasing the number of SCFAs-producing Bifidobacterium and Roseburia. Subsequently, the short-chain fatty acid (SCFA) content was examined and WC10 significantly increased acetate, propionate, and butyrate production. Additionally, metabolomic analysis also showed that WC10 reversed the antibiotic interference with major metabolic pathways. These findings provide a solid scientific basis for the future application of W. coagulans WC10 in the treatment of AAD.
Collapse
Affiliation(s)
- Zengbo Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
- Food College, Northeast Agricultural University, Harbin, 150030, China
| | - Zengtao Guo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
- Food College, Northeast Agricultural University, Harbin, 150030, China
| | - Libo Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
- Food College, Northeast Agricultural University, Harbin, 150030, China
| | - Daxi Ren
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hang Zu
- Heilongjiang Ubert Dairy Co., Heilongjiang, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.
- Food College, Northeast Agricultural University, Harbin, 150030, China.
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.
- Food College, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
10
|
Ballanti M, Antonetti L, Mavilio M, Casagrande V, Moscatelli A, Pietrucci D, Teofani A, Internò C, Cardellini M, Paoluzi O, Monteleone G, Lefebvre P, Staels B, Mingrone G, Menghini R, Federici M. Decreased circulating IPA levels identify subjects with metabolic comorbidities: A multi-omics study. Pharmacol Res 2024; 204:107207. [PMID: 38734193 DOI: 10.1016/j.phrs.2024.107207] [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: 03/13/2024] [Revised: 05/05/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
In recent years several experimental observations demonstrated that the gut microbiome plays a role in regulating positively or negatively metabolic homeostasis. Indole-3-propionic acid (IPA), a Tryptophan catabolic product mainly produced by C. Sporogenes, has been recently shown to exert either favorable or unfavorable effects in the context of metabolic and cardiovascular diseases. We performed a study to delineate clinical and multiomics characteristics of human subjects characterized by low and high IPA levels. Subjects with low IPA blood levels showed insulin resistance, overweight, low-grade inflammation, and features of metabolic syndrome compared to those with high IPA. Metabolomics analysis revealed that IPA was negatively correlated with leucine, isoleucine, and valine metabolism. Transcriptomics analysis in colon tissue revealed the enrichment of several signaling, regulatory, and metabolic processes. Metagenomics revealed several OTU of ruminococcus, alistipes, blautia, butyrivibrio and akkermansia were significantly enriched in highIPA group while in lowIPA group Escherichia-Shigella, megasphera, and Desulfovibrio genus were more abundant. Next, we tested the hypothesis that treatment with IPA in a mouse model may recapitulate the observations of human subjects, at least in part. We found that a short treatment with IPA (4 days at 20/mg/kg) improved glucose tolerance and Akt phosphorylation in the skeletal muscle level, while regulating blood BCAA levels and gene expression in colon tissue, all consistent with results observed in human subjects stratified for IPA levels. Our results suggest that treatment with IPA may be considered a potential strategy to improve insulin resistance in subjects with dysbiosis.
Collapse
Affiliation(s)
- Marta Ballanti
- Center for Atherosclerosis and Internal Medicine Unit, Policlinico Tor Vergata University Hospital, Via Oxford 81, Rome 00133, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Lorenzo Antonetti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Maria Mavilio
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Viviana Casagrande
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Alessandro Moscatelli
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy; Laboratory of Neuromotor Physiology, Santa Lucia Foundation IRCCS, Rome, 00179, Italy
| | - Daniele Pietrucci
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Adelaide Teofani
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Chiara Internò
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Marina Cardellini
- Center for Atherosclerosis and Internal Medicine Unit, Policlinico Tor Vergata University Hospital, Via Oxford 81, Rome 00133, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Omero Paoluzi
- Unit of Gastroenterology, Policlinico Tor Vergata University Hospital, Via Oxford 81, 00133 Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy; Unit of Gastroenterology, Policlinico Tor Vergata University Hospital, Via Oxford 81, 00133 Rome, Italy
| | - Philippe Lefebvre
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 EGID, Lille France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 EGID, Lille France
| | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; Diabetes and Nutritional Sciences, Hodgkin Building, Guy's Campus, King's College London, London WC2R 2LS, UK
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Massimo Federici
- Center for Atherosclerosis and Internal Medicine Unit, Policlinico Tor Vergata University Hospital, Via Oxford 81, Rome 00133, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy.
| |
Collapse
|
11
|
Jin Q, Zhang C, Chen R, Jiang L, Li H, Wu P, Li L. Quinic acid regulated TMA/TMAO-related lipid metabolism and vascular endothelial function through gut microbiota to inhibit atherosclerotic. J Transl Med 2024; 22:352. [PMID: 38622667 PMCID: PMC11017595 DOI: 10.1186/s12967-024-05120-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/20/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Quinic acid (QA) and its derivatives have good lipid-lowering and hepatoprotective functions, but their role in atherosclerosis remains unknown. This study attempted to investigate the mechanism of QA on atherogenesis in Apoe-/- mice induced by HFD. METHODS HE staining and oil red O staining were used to observe the pathology. The PCSK9, Mac-3 and SM22a expressions were detected by IHC. Cholesterol, HMGB1, TIMP-1 and CXCL13 levels were measured by biochemical and ELISA. Lipid metabolism and the HMGB1-SREBP2-SR-BI pathway were detected by PCR and WB. 16 S and metabolomics were used to detect gut microbiota and serum metabolites. RESULTS QA or low-frequency ABX inhibited weight gain and aortic tissue atherogenesis in HFD-induced Apoe-/- mice. QA inhibited the increase of cholesterol, TMA, TMAO, CXCL13, TIMP-1 and HMGB1 levels in peripheral blood of Apoe-/- mice induced by HFD. Meanwhile, QA or low-frequency ABX treatment inhibited the expression of CAV-1, ABCA1, Mac-3 and SM22α, and promoted the expression of SREBP-1 and LXR in the vascular tissues of HFD-induced Apoe-/- mice. QA reduced Streptococcus_danieliae abundance, and promoted Lactobacillus_intestinalis and Ileibacterium_valens abundance in HFD-induced Apoe-/- mice. QA altered serum galactose metabolism, promoted SREBP-2 and LDLR, inhibited IDOL, FMO3 and PCSK9 expression in liver of HFD-induced Apoe-/- mice. The combined treatment of QA and low-frequency ABX regulated microbe-related Glycoursodeoxycholic acid and GLYCOCHENODEOXYCHOLATE metabolism in HFD-induced Apoe-/- mice. QA inhibited TMAO or LDL-induced HCAECs damage and HMGB1/SREBP2 axis dysfunction, which was reversed by HMGB1 overexpression. CONCLUSIONS QA regulated the gut-liver lipid metabolism and chronic vascular inflammation of TMA/TMAO through gut microbiota to inhibit the atherogenesis in Apoe-/- mice, and the mechanism may be related to the HMGB1/SREBP2 pathway.
Collapse
Affiliation(s)
- Qiao Jin
- Department of Cardiovascular Medicine, Hengyang Medical School, The Changsha central Affiliated Hospital, University of South China, Changsha, Hunan, 410004, China
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, 410013, China
| | - Chiyuan Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ran Chen
- Department of Cardiovascular Medicine, Hengyang Medical School, The Changsha central Affiliated Hospital, University of South China, Changsha, Hunan, 410004, China
| | - Luping Jiang
- Department of Cardiovascular Medicine, Hengyang Medical School, The Changsha central Affiliated Hospital, University of South China, Changsha, Hunan, 410004, China
| | - Hongli Li
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Pengcui Wu
- Department of Cardiovascular Medicine, Hengyang Medical School, The Changsha central Affiliated Hospital, University of South China, Changsha, Hunan, 410004, China.
| | - Liang Li
- Department of Cardiovascular Medicine, Hengyang Medical School, The Changsha central Affiliated Hospital, University of South China, Changsha, Hunan, 410004, China.
| |
Collapse
|
12
|
Muller E, Shiryan I, Borenstein E. Multi-omic integration of microbiome data for identifying disease-associated modules. Nat Commun 2024; 15:2621. [PMID: 38521774 PMCID: PMC10960825 DOI: 10.1038/s41467-024-46888-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
Multi-omic studies of the human gut microbiome are crucial for understanding its role in disease across multiple functional layers. Nevertheless, integrating and analyzing such complex datasets poses significant challenges. Most notably, current analysis methods often yield extensive lists of disease-associated features (e.g., species, pathways, or metabolites), without capturing the multi-layered structure of the data. Here, we address this challenge by introducing "MintTea", an intermediate integration-based approach combining canonical correlation analysis extensions, consensus analysis, and an evaluation protocol. MintTea identifies "disease-associated multi-omic modules", comprising features from multiple omics that shift in concord and that collectively associate with the disease. Applied to diverse cohorts, MintTea captures modules with high predictive power, significant cross-omic correlations, and alignment with known microbiome-disease associations. For example, analyzing samples from a metabolic syndrome study, MintTea identifies a module with serum glutamate- and TCA cycle-related metabolites, along with bacterial species linked to insulin resistance. In another dataset, MintTea identifies a module associated with late-stage colorectal cancer, including Peptostreptococcus and Gemella species and fecal amino acids, in line with these species' metabolic activity and their coordinated gradual increase with cancer development. This work demonstrates the potential of advanced integration methods in generating systems-level, multifaceted hypotheses underlying microbiome-disease interactions.
Collapse
Affiliation(s)
- Efrat Muller
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Shiryan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Elhanan Borenstein
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel.
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
13
|
Hemmati M, Kashanipoor S, Mazaheri P, Alibabaei F, Babaeizad A, Asli S, Mohammadi S, Gorgin AH, Ghods K, Yousefi B, Eslami M. Importance of gut microbiota metabolites in the development of cardiovascular diseases (CVD). Life Sci 2023; 329:121947. [PMID: 37463653 DOI: 10.1016/j.lfs.2023.121947] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023]
Abstract
Cardiovascular disease (CVD) remains the most common cause of death worldwide and has become a public health concern. The proven notable risk factors for CVD are atherosclerosis, hypertension, diabetes, dyslipidemia, inflammation, and some genetic defects. However, research has shown a correlation between metabolic health, gut microbiota, and dietary risk factors. The gut microbiota makes an important contribution to human functional metabolic pathways by contributing enzymes that are not encoded by the human genome, for instance, the breakdown of polysaccharides, polyphenols and vitamins synthesis. TMAO and SCFAs, human gut microbiota compounds, have respective immunomodulatory and pro-inflammatory effects. Choline and l-carnitine are abundant in high-fat diets and are transformed into TMA by gut bacteria. The liver's phase of metabolism then changes TMA into TMAO. In turn, TMAO promotes the activation of macrophages, damages vascular endothelium, and results in CVD-however, dysbiosis decreases SCFAs and bile acids, which raises intestinal permeability. Congestion in the portal vein, a drop in cardiac output, a reduction in intestinal perfusion, and intestinal leakage are all caused by heart failure. These factors induce systemic inflammation by increasing intestinal leakage. By raising CRP and pro-inflammatory reactions, human gut dysbiosis and elevated TMAO levels promote the development of arterial plaque, hasten the beginning of atherosclerosis, and raise the risk of CAD. A healthy symbiosis between the gut microbiota and host is a key factor in shaping the biochemical profile of the diet, therefore which are crucial for maintaining the intestinal epithelial barrier, growing mucosa, reducing inflammation, and controlling blood pressure.
Collapse
Affiliation(s)
- Maryam Hemmati
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Payman Mazaheri
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Farnaz Alibabaei
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Shima Asli
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sina Mohammadi
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Amir Hosein Gorgin
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Kamran Ghods
- Social Determinants of Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
14
|
Witkowska AM, Salem JE. Pharmacological and Nutritional Modulation of Metabolome and Metagenome in Cardiometabolic Disorders. Biomolecules 2023; 13:1340. [PMID: 37759740 PMCID: PMC10526920 DOI: 10.3390/biom13091340] [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: 07/24/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiometabolic disorders are major causes of morbidity and mortality worldwide. A growing body of research indicates that the gut microbiota, whether it interacts favorably or not, plays an important role in host metabolism. Elucidating metabolic pathways may be crucial in preventing and treating cardiometabolic diseases, and omics methods are key to studying the interaction between the fecal microbiota and host metabolism. This review summarizes available studies that combine metabolomic and metagenomic approaches to describe the effects of drugs, diet, nutrients, and specific foods on cardiometabolic health and to identify potential targets for future research.
Collapse
Affiliation(s)
- Anna Maria Witkowska
- Department of Food Biotechnology, Faculty of Health Sciences, Medical University of Bialystok, Szpitalna 37, 15-295 Białystok, Poland
| | - Joe-Elie Salem
- Department of Pharmacology, Pitié-Salpêtrière Hospital, Institut National de la Santé et de la Recherche Médicale (INSERM), Clinical Investigation Center (CIC-1901), Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne Université, 75013 Paris, France;
| |
Collapse
|
15
|
Xu K, Saaoud F, Shao Y, Lu Y, Wu S, Zhao H, Chen K, Vazquez-Padron R, Jiang X, Wang H, Yang X. Early hyperlipidemia triggers metabolomic reprogramming with increased SAH, increased acetyl-CoA-cholesterol synthesis, and decreased glycolysis. Redox Biol 2023; 64:102771. [PMID: 37364513 PMCID: PMC10310484 DOI: 10.1016/j.redox.2023.102771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
To identify metabolomic reprogramming in early hyperlipidemia, unbiased metabolome was screened in four tissues from ApoE-/- mice fed with high fat diet (HFD) for 3 weeks. 30, 122, 67, and 97 metabolites in the aorta, heart, liver, and plasma, respectively, were upregulated. 9 upregulated metabolites were uremic toxins, and 13 metabolites, including palmitate, promoted a trained immunity with increased syntheses of acetyl-CoA and cholesterol, increased S-adenosylhomocysteine (SAH) and hypomethylation and decreased glycolysis. The cross-omics analysis found upregulation of 11 metabolite synthetases in ApoE‾/‾ aorta, which promote ROS, cholesterol biosynthesis, and inflammation. Statistical correlation of 12 upregulated metabolites with 37 gene upregulations in ApoE‾/‾ aorta indicated 9 upregulated new metabolites to be proatherogenic. Antioxidant transcription factor NRF2-/- transcriptome analysis indicated that NRF2 suppresses trained immunity-metabolomic reprogramming. Our results have provided novel insights on metabolomic reprogramming in multiple tissues in early hyperlipidemia oriented toward three co-existed new types of trained immunity.
Collapse
Affiliation(s)
- Keman Xu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ying Shao
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Yifan Lu
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Sheng Wu
- Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Huaqing Zhao
- Medical Education and Data Science, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Kaifu Chen
- Computational Biology Program, Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Roberto Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33125, USA
| | - Xiaohua Jiang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA; Metabolic Disease Research, Thrombosis Research, Departments of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
16
|
Baranwal G, Goodlett BL, Arenaz CM, Creed HA, Navaneethabalakrishnan S, Rutkowski JM, Alaniz RC, Mitchell BM. Indole Propionic Acid Increases T Regulatory Cells and Decreases T Helper 17 Cells and Blood Pressure in Mice with Salt-Sensitive Hypertension. Int J Mol Sci 2023; 24:9192. [PMID: 37298145 PMCID: PMC10252743 DOI: 10.3390/ijms24119192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
Hypertension affects over a billion adults worldwide and is a major risk factor for cardiovascular disease. Studies have reported that the microbiota and its metabolites regulate hypertension pathophysiology. Recently, tryptophan metabolites have been identified to contribute to and inhibit the progression of metabolic disorders and cardiovascular diseases, including hypertension. Indole propionic acid (IPA) is a tryptophan metabolite with reported protective effects in neurodegenerative and cardiovascular diseases; however, its involvement in renal immunomodulation and sodium handling in hypertension is unknown. In the current study, targeted metabolomic analysis revealed decreased serum and fecal IPA levels in mice with L-arginine methyl ester hydrochloride (L-NAME)/high salt diet-induced hypertension (LSHTN) compared to normotensive control mice. Additionally, kidneys from LSHTN mice had increased T helper 17 (Th17) cells and decreased T regulatory (Treg) cells. Dietary IPA supplementation in LSHTN mice for 3 weeks resulted in decreased systolic blood pressure, along with increased total 24 h and fractional sodium excretion. Kidney immunophenotyping demonstrated decreased Th17 cells and a trend toward increased Treg cells in IPA-supplemented LSHTN mice. In vitro, naïve T cells from control mice were skewed into Th17 or Treg cells. The presence of IPA decreased Th17 cells and increased Treg cells after 3 days. These results identify a direct role for IPA in attenuating renal Th17 cells and increasing Treg cells, leading to improved sodium handling and decreased blood pressure. IPA may be a potential metabolite-based therapeutic option for hypertension.
Collapse
Affiliation(s)
- Gaurav Baranwal
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807, USA (B.L.G.)
| | - Bethany L. Goodlett
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807, USA (B.L.G.)
| | - Cristina M. Arenaz
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807, USA (B.L.G.)
| | - Heidi A. Creed
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807, USA (B.L.G.)
| | | | - Joseph M. Rutkowski
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807, USA (B.L.G.)
| | - Robert C. Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University School of Medicine, Bryan, TX 77807, USA
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX 77807, USA (B.L.G.)
| |
Collapse
|
17
|
The Tryptophan and Kynurenine Pathway Involved in the Development of Immune-Related Diseases. Int J Mol Sci 2023; 24:ijms24065742. [PMID: 36982811 PMCID: PMC10051340 DOI: 10.3390/ijms24065742] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
The tryptophan and kynurenine pathway is well-known to play an important role in nervous, endocrine, and immune systems, as well as in the development of inflammatory diseases. It has been documented that some kynurenine metabolites are considered to have anti-oxidative, anti-inflammatory, and/or neuroprotective properties. Importantly, many of these kynurenine metabolites may possess immune-regulatory properties that could alleviate the inflammation response. The abnormal activation of the tryptophan and kynurenine pathway might be involved in the pathophysiological process of various immune-related diseases, such as inflammatory bowel disease, cardiovascular disease, osteoporosis, and/or polycystic ovary syndrome. Interestingly, kynurenine metabolites may be involved in the brain memory system and/or intricate immunity via the modulation of glial function. In the further deliberation of this concept with engram, the roles of gut microbiota could lead to the development of remarkable treatments for the prevention of and/or the therapeutics for various intractable immune-related diseases.
Collapse
|
18
|
Li Y, Yang S, Jin X, Li D, Lu J, Wang X, Wu M. Mitochondria as novel mediators linking gut microbiota to atherosclerosis that is ameliorated by herbal medicine: A review. Front Pharmacol 2023; 14:1082817. [PMID: 36733506 PMCID: PMC9886688 DOI: 10.3389/fphar.2023.1082817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Atherosclerosis (AS) is the main cause of cardiovascular disease (CVD) and is characterized by endothelial damage, lipid deposition, and chronic inflammation. Gut microbiota plays an important role in the occurrence and development of AS by regulating host metabolism and immunity. As human mitochondria evolved from primordial bacteria have homologous characteristics, they are attacked by microbial pathogens as target organelles, thus contributing to energy metabolism disorders, oxidative stress, and apoptosis. Therefore, mitochondria may be a key mediator of intestinal microbiota disorders and AS aggravation. Microbial metabolites, such as short-chain fatty acids, trimethylamine, hydrogen sulfide, and bile acids, also affect mitochondrial function, including mtDNA mutation, oxidative stress, and mitophagy, promoting low-grade inflammation. This further damages cellular homeostasis and the balance of innate immunity, aggravating AS. Herbal medicines and their monomers can effectively ameliorate the intestinal flora and their metabolites, improve mitochondrial function, and inhibit atherosclerotic plaques. This review focuses on the interaction between gut microbiota and mitochondria in AS and explores a therapeutic strategy for restoring mitochondrial function and intestinal microbiota disorders using herbal medicines, aiming to provide new insights for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yujuan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Beijing University of Chinese Medicine, Beijing, China
| | - Xinyue Wang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Min Wu,
| |
Collapse
|
19
|
Advances in Lactobacillus Restoration for β-Lactam Antibiotic-Induced Dysbiosis: A System Review in Intestinal Microbiota and Immune Homeostasis. Microorganisms 2023; 11:microorganisms11010179. [PMID: 36677471 PMCID: PMC9861108 DOI: 10.3390/microorganisms11010179] [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: 11/02/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
A balanced gut microbiota and their metabolites are necessary for the maintenance of the host's health. The antibiotic-induced dysbiosis can cause the disturbance of the microbial community, influence the immune homeostasis and induce susceptibility to metabolic- or immune-mediated disorders and diseases. The Lactobacillus and their metabolites or components affect the function of the host's immune system and result in microbiota-mediated restoration. Recent data have indicated that, by altering the composition and functions of gut microbiota, antibiotic exposure can also lead to a number of specific pathologies, hence, understanding the potential mechanisms of the interactions between gut microbiota dysbiosis and immunological homeostasis is very important. The Lactobacillus strategies for detecting the associations between the restoration of the relatively imbalanced microbiome and gut diseases are provided in this discussion. In this review, we discuss the recently discovered connections between microbial communities and metabolites in the Lactobacillus treatment of β-lactam antibiotic-induced dysbiosis, and establish the relationship between commensal bacteria and host immunity under this imbalanced homeostasis of the gut microbiota.
Collapse
|
20
|
Salazar J, Morillo V, Suárez MK, Castro A, Ramírez P, Rojas M, Añez R, D’Marco L, Chacín-González M, Bermudez V. Role of Gut Microbiome in Atherosclerosis: Molecular and Therapeutic Aspects. Curr Cardiol Rev 2023; 19:e020223213408. [PMID: 36733248 PMCID: PMC10494273 DOI: 10.2174/1573403x19666230202164524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is one of the most relevant and prevalent cardiovascular diseases of our time. It is one of the pathological entities that increases the morbidity and mortality index in the adult population. Pathophysiological connections have been observed between atherosclerosis and the gut microbiome (GM), represented by a group of microorganisms that are present in the gut. These microorganisms are vital for metabolic homeostasis in humans. Recently, direct and indirect mechanisms through which GM can affect the development of atherosclerosis have been studied. This has led to research into the possible modulation of GM and metabolites as a new target in the prevention and treatment of atherosclerosis. The goal of this review is to analyze the physiopathological mechanisms linking GM and atherosclerosis that have been described so far. We also aim to summarize the recent studies that propose GM as a potential target in atherosclerosis management.
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Valery Morillo
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María K Suárez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ana Castro
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Paola Ramírez
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Disease Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición. Hospital General Universitario Gregorio Marañón, Madrid, España
| | - Luis D’Marco
- Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, 46115, Spain
| | | | - Valmore Bermudez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| |
Collapse
|
21
|
Kappel BA, De Angelis L, Puetz A, Ballanti M, Menghini R, Marx N, Federici M. Antibiotic-induced gut microbiota depletion exacerbates host hypercholesterolemia. Pharmacol Res 2023; 187:106570. [PMID: 36423788 DOI: 10.1016/j.phrs.2022.106570] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/06/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022]
Abstract
Hypercholesterolemia is a major driver of atherosclerosis, thus contributing to high morbidity and mortality worldwide. Gut microbiota have been identified as modulator of blood lipids including cholesterol levels. Few studies have already linked certain bacteria and microbial mechanisms to host cholesterol. However, in particular mouse models revealed conflicting results depending on genetics and experimental protocol. To gain further insights into the relationship between intestinal bacteria and host cholesterol metabolism, we first performed fecal 16S rRNA targeted metagenomic sequencing in a human cohort (n = 24) naïve for cholesterol lowering drugs. Here, we show alterations in the gut microbiota composition of hypercholesterolemic patients with depletion of Bifidobacteria, expansion of Clostridia and increased Firmicutes/Bacteroidetes ratio. To test whether pharmacological intervention in gut microbiota impacts host serum levels of cholesterol, we treated hypercholesterolemic Apolipoprotein E knockout with oral largely non-absorbable antibiotics. Antibiotics increased serum cholesterol, but only when mice were fed normal chow diet and cholesterol was measured in the random fed state. These elevations in cholesterol already occurred few days after treatment initiation and were reversible after stopping antibiotics with re-acquisition of intestinal bacteria. Gene expression analyses pointed to increased intestinal cholesterol uptake mediated by antibiotics in the fed state. Non-targeted serum metabolomics suggested that diminished plant sterol levels and reduced bile acid cycling were involved microbial mechanisms. In conclusion, our work further enlightens the link between gut microbiota and host cholesterol metabolism. Pharmacological disruption of the gut flora by antibiotics was able to exacerbate serum cholesterol and may impact cardiovascular disease.
Collapse
Affiliation(s)
- Ben A Kappel
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Lorenzo De Angelis
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andreas Puetz
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Marta Ballanti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nikolaus Marx
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
22
|
Liao XX, Wu XY, Zhou YL, Li JJ, Wen YL, Zhou JJ. Gut microbiome metabolites as key actors in atherosclerosis co-depression disease. Front Microbiol 2022; 13:988643. [PMID: 36439791 PMCID: PMC9686300 DOI: 10.3389/fmicb.2022.988643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/24/2022] [Indexed: 02/26/2024] Open
Abstract
Cardiovascular diseases, mainly characterized by atherosclerosis (AS), and depression have a high comorbidity rate. However, previous studies have been conducted under a single disease, and there is a lack of studies in comorbid states to explore the commonalities in the pathogenesis of both diseases. Modern high-throughput technologies have made it clear that the gut microbiome can affect the development of the host's own disorders and have shown that their metabolites are crucial to the pathophysiology of AS and depression. The aim of this review is to summarize the current important findings on the role of gut microbiome metabolites such as pathogen-associated molecular patterns, bile acids, tryptophan metabolites, short-chain fatty acids, and trimethylamine N -oxide in depression and AS disease, with the aim of identifying potential biological targets for the early diagnosis and treatment of AS co-depression disorders.
Collapse
Affiliation(s)
- Xing-Xing Liao
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Xiao-Yun Wu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yu-Long Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Jia-Jun Li
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - You-Liang Wen
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Jun-Jie Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
23
|
del Castillo-Izquierdo Á, Moreno-Navarrete JM, Latorre J, Arnoriaga-Rodríguez M, Ballanti M, Monteleone G, Alessandro Paoluzi O, Mingrone G, Puig J, Ramos R, Garre-Olmo J, Jové M, Pamplona R, Portero-Otín M, Sol J, Lefebvre P, Staels B, Federici M, Fernández-Real JM, Mayneris-Perxachs J. DPP9 as a Potential Novel Mediator in Gastrointestinal Virus Infection. Antioxidants (Basel) 2022; 11:2177. [PMID: 36358551 PMCID: PMC9686780 DOI: 10.3390/antiox11112177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 01/02/2025] Open
Abstract
Dipeptidyl peptidase 9 (DPP9) is a member of the dipeptidyl peptidase IV family. Inhibition of DPP9 has recently been shown to activate the nucleotide-binding domain leucine-rich repeat 1 (NLRP1) inflammasome. NLRP1 is known to bind nucleic acids with high affinity and directly interact with double stranded RNA, which plays a key role in viral replication. DPP9 has also recently emerged as a key gene related to lung-inflammation in critical SARS-CoV-2 infection. Importantly, DPP9 activity is strongly dependent on the oxidative status. Here, we explored the potential role of DPP9 in the gastrointestinal tract. We performed transcriptomics analyses of colon (microarray, n = 37) and jejunal (RNA sequencing, n = 31) biopsies from two independent cohorts as well as plasma metabolomics analyses in two independent cohorts (n = 37 and n = 795). The expression of DPP9 in the jejunum, colon, and blood was significantly associated with circulating biomarkers of oxidative stress (uric acid, bilirubin). It was also associated positively with the expression of transcription factors (NRF-2) and genes (SOD, CAT, GPX) encoding for antioxidant enzymes, but negatively with that of genes (XDH, NOX) and transcription factors (NF-KB) involved in ROS-generating enzymes. Gene co-expression patterns associated with DPP9 identified several genes participating in antiviral pathways in both tissues. Notably, DPP9 expression in the colon and plasma was strongly positively associated with several circulating nucleotide catabolites (hypoxanthine, uric acid, 3-ureidopropionic acid) with important roles in the generation of ROS and viral infection, as well as other metabolites related to oxidative stress (Resolvin D1, glutamate-containing dipeptides). Gene-drug enrichment analyses identified artenimol, puromycin, anisomycin, 3-phenyllactic acid, and linezolid as the most promising drugs targeting these DPP9-associated genes. We have identified a novel potential pathogenic mechanism of viral infection in the digestive tract and promising existing drugs that can be repositioned against viral infection.
Collapse
Affiliation(s)
- Ángela del Castillo-Izquierdo
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - Jessica Latorre
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Arnoriaga-Rodríguez
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - Marta Ballanti
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Center for Atherosclerosis, Policlinico Tor Vergata, 00133 Rome, Italy
| | | | | | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Diabetes and Nutritional Sciences, Hodgkin Building, Guy’s Campus, King’s College London, London WC2R 2LS, UK
| | - Josep Puig
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
- Department of Radiology (IDI), Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain
| | - Rafael Ramos
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
- Vascular Health Research Group of Girona (ISV-Girona), Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), 08007 Barcelona, Spain
- Primary Care Services, Catalan Institute of Health (ICS), 17007 Girona, Spain
| | - Josep Garre-Olmo
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
- Vascular Health Research Group of Girona (ISV-Girona), Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), 08007 Barcelona, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), 25198 Lleida, Spain
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), 25198 Lleida, Spain
| | - Joaquim Sol
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), 25198 Lleida, Spain
| | - Philippe Lefebvre
- Institut Pasteur de Lille, Université Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Bart Staels
- Institut Pasteur de Lille, Université Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Center for Atherosclerosis, Policlinico Tor Vergata, 00133 Rome, Italy
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004 Girona, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, 17007 Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
24
|
Francisco AJ. Helicobacter Pylori Infection Induces Intestinal Dysbiosis That Could Be Related to the Onset of Atherosclerosis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9943158. [PMID: 36317116 PMCID: PMC9617700 DOI: 10.1155/2022/9943158] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022]
Abstract
Cardiovascular diseases represent one of the first causes of death around the world, and atherosclerosis is one of the first steps in the development of them. Although these problems occur mainly in elderly, the incidence in younger people is being reported, and an undetermined portion of patients without the classic risk factors develop subclinical atherosclerosis at earlier stages of life. Recently, both the H. pylori infection and the intestinal microbiota have been linked to atherosclerosis. The mechanisms behind those associations are poorly understood, but some of the proposed explanations are (a) the effect of the chronic systemic inflammation induced by H. pylori, (b) a direct action over the endothelial cells by the cytotoxin associated gene A protein, and (c) alterations of the lipid metabolism and endothelial dysfunction induced by H. pylori infection. Regarding the microbiota, several studies show that induction of atherosclerosis is related to high levels of Trimethylamine N-oxide. In this review, we present the information published about the effects of H. pylori over the intestinal microbiota and their relationship with atherosclerosis and propose a hypothesis to explain the nature of these associations. If H. pylori contributes to atherosclerosis, then interventions for eradication and restoration of the gut microbiota at early stages could represent a way to prevent disease progression.
Collapse
Affiliation(s)
- Avilés-Jiménez Francisco
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Pediatría. Centro Médico Nacional Siglo XXI. IMSS, Ciudad de México, Mexico
| |
Collapse
|
25
|
Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. MOLECULAR BIOMEDICINE 2022; 3:30. [PMID: 36219347 PMCID: PMC9554112 DOI: 10.1186/s43556-022-00091-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota is critical to human health, such as digesting nutrients, forming the intestinal epithelial barrier, regulating immune function, producing vitamins and hormones, and producing metabolites to interact with the host. Meanwhile, increasing evidence indicates that the gut microbiota has a strong correlation with the occurrence, progression and treatment of cardiovascular diseases (CVDs). In patients with CVDs and corresponding risk factors, the composition and ratio of gut microbiota have significant differences compared with their healthy counterparts. Therefore, gut microbiota dysbiosis, gut microbiota-generated metabolites, and the related signaling pathway may serve as explanations for some of the mechanisms about the occurrence and development of CVDs. Several studies have also demonstrated that many traditional and latest therapeutic treatments of CVDs are associated with the gut microbiota and its generated metabolites and related signaling pathways. Given that information, we summarized the latest advances in the current research regarding the effect of gut microbiota on health, the main cardiovascular risk factors, and CVDs, highlighted the roles and mechanisms of several metabolites, and introduced corresponding promising treatments for CVDs regarding the gut microbiota. Therefore, this review mainly focuses on exploring the role of gut microbiota related metabolites and their therapeutic potential in CVDs, which may eventually provide better solutions in the development of therapeutic treatment as well as the prevention of CVDs.
Collapse
Affiliation(s)
- Lu Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Shiqi Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Qing Zhang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chengqi He
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chenying Fu
- grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,grid.412901.f0000 0004 1770 1022Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Quan Wei
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
26
|
Paeslack N, Mimmler M, Becker S, Gao Z, Khuu MP, Mann A, Malinarich F, Regen T, Reinhardt C. Microbiota-derived tryptophan metabolites in vascular inflammation and cardiovascular disease. Amino Acids 2022; 54:1339-1356. [PMID: 35451695 PMCID: PMC9641817 DOI: 10.1007/s00726-022-03161-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/27/2022] [Indexed: 12/17/2022]
Abstract
The essential amino acid tryptophan (Trp) is metabolized by gut commensals, yielding in compounds that affect innate immune cell functions directly, but also acting on the aryl hydrocarbon receptor (AHR), thus regulating the maintenance of group 3 innate lymphoid cells (ILCs), promoting T helper 17 (TH17) cell differentiation, and interleukin-22 production. In addition, microbiota-derived Trp metabolites have direct effects on the vascular endothelium, thus influencing the development of vascular inflammatory phenotypes. Indoxyl sulfate was demonstrated to promote vascular inflammation, whereas indole-3-propionic acid and indole-3-aldehyde had protective roles. Furthermore, there is increasing evidence for a contributory role of microbiota-derived indole-derivatives in blood pressure regulation and hypertension. Interestingly, there are indications for a role of the kynurenine pathway in atherosclerotic lesion development. Here, we provide an overview on the emerging role of gut commensals in the modulation of Trp metabolism and its influence in cardiovascular disease development.
Collapse
Affiliation(s)
- Nadja Paeslack
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Maximilian Mimmler
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Stefanie Becker
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Zhenling Gao
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - My Phung Khuu
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Amrit Mann
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Frano Malinarich
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
27
|
Li Z, Liu K, Zhao J, Yang L, Chen G, Liu A, Wang Q, Wang S, Li X, Cao H, Tao F, Zhang D. Antibiotics in elderly Chinese population and their relations with hypertension and pulse pressure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:67026-67045. [PMID: 35513617 DOI: 10.1007/s11356-022-20613-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/30/2022] [Indexed: 06/14/2023]
Abstract
Although antibiotic exposure in the general population has been well documented by a biomonitoring approach, epidemiologic data on the relationships between urinary antibiotic burden in the elderly with blood pressure (BP) are still lacking. The current study revealed thirty-four antibiotics in urine specimens from 990 elderly patients in Lu'an City, China, with detection frequencies ranging from 0.2 to 35.5%. Among the elderly, the prevalence of hypertension was 72.0%, and 12 antibiotics were detected in more than 10% of individuals with hypertension. The elderly with hypertension had the maximum daily exposure (5450.45 μg/kg/day) to fluoroquinolones (FQs). Multiple linear regression analyses revealed significant associations of BP and pulse pressure (PP) with exposure to specific antibiotics. The estimated β values (95% confidence interval) of associations with systolic blood pressure (SBP) in the right arm were 4.42 (1.15, 7.69) for FQs, 4.26 (0.52, 8.01) for the preferred as human antibiotics (PHAs), and 3.48 (0.20, 6.77) for the mixtures (FQs + tetracyclines [TCs] (tertile 3 vs. tertile 1)), respectively. Increased concentrations of TCs were associated with decreased diastolic BP (DBP; tertile 3: -1.75 [-3.39, -0.12]) for the right arm. Higher levels of FQs (tertile 3: 4.28 [1.02, 7.54]), PHAs (tertile 3: 4.25 [0.49, 8.01]), and FQs + TCs (tertile 3: 3.99 [0.71, 7.26]) were associated with increased SBP, and an increase in DBP for FQs (tertile 3: 1.82 [0.22, 3.42]) was shown in the left arm. Also, higher urinary concentrations of FQs (tertile 3: 3.18 [0.53, 5.82]), PHAs (tertile 3: 3.42 [0.40, 6.45]), and FQs + TCs (tertile 3: 3.06 [0.40, 5.72]) were related to increased PP, whereas a decline in PP for TCs (tertile 2: -2.93 [-5.60, -0.25]) in the right arm. And increased concentrations of penicillin V (tertile 3: 5.31 [1.53, 9.10]) and FQs + TCs (tertile 3: 2.84 [0.19, 5.49]) were related to higher PP in the left arm. By utilizing restricted cubic splines, our current study revealed a potential nonlinear dose-response association between FQ exposure and hypertension risk. In conclusion, this investigation is the first to present antibiotic exposure using a biomonitoring approach, and informs understanding of impacts of antibiotic residues, as emerging hazardous pollutants, on the hypertension risk in the elderly.
Collapse
Affiliation(s)
- Zhenkun Li
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jianing Zhao
- The Fourth Affiliated Hospital of Anhui Medical University, Huaihai Road, Hefei, 230012, Anhui, China
| | - Linsheng Yang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Guimei Chen
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Annuo Liu
- School of Nursing, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Qunan Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Sufang Wang
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xiude Li
- Lu'an Center of Disease Control and Prevention, Lu'an, 237000, Anhui, China
| | - Hongjuan Cao
- Lu'an Center of Disease Control and Prevention, Lu'an, 237000, Anhui, China
| | - Fangbiao Tao
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Dongmei Zhang
- School of Health Management, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
28
|
The Gut Microbiota and Vascular Aging: A State-of-the-Art and Systematic Review of the Literature. J Clin Med 2022; 11:jcm11123557. [PMID: 35743626 PMCID: PMC9224769 DOI: 10.3390/jcm11123557] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota is a critical regulator of human physiology, deleterious changes to its composition and function (dysbiosis) have been linked to the development and progression of cardiovascular diseases. Vascular ageing (VA) is a process of progressive stiffening of the arterial tree associated with arterial wall remodeling, which can precede hypertension and organ damage, and is associated with cardiovascular risk. Arterial stiffness has become the preferred marker of VA. In our systematic review, we found an association between gut microbiota composition and arterial stiffness, with two patterns, in most animal and human studies: a direct correlation between arterial stiffness and abundances of bacteria associated with altered gut permeability and inflammation; an inverse relationship between arterial stiffness, microbiota diversity, and abundances of bacteria associated with most fit microbiota composition. Interventional studies were able to show a stable link between microbiota modification and arterial stiffness only in animals. None of the human interventional trials was able to demonstrate this relationship, and very few adjusted the analyses for determinants of arterial stiffness. We observed a lack of large randomized interventional trials in humans that test the role of gut microbiota modifications on arterial stiffness, and take into account BP and hemodynamic alterations.
Collapse
|
29
|
Interactions between Tryptophan Metabolism, the Gut Microbiome and the Immune System as Potential Drivers of Non-Alcoholic Fatty Liver Disease (NAFLD) and Metabolic Diseases. Metabolites 2022; 12:metabo12060514. [PMID: 35736447 PMCID: PMC9227929 DOI: 10.3390/metabo12060514] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing and therefore is its burden of disease as NALFD is a risk factor for cirrhosis and is associated with other metabolic conditions such as type II diabetes, obesity, dyslipidaemia and atherosclerosis. Linking these cardiometabolic diseases is a state of low-grade inflammation, with higher cytokines and c-reactive protein levels found in individuals with NAFLD, obesity and type II diabetes. A possible therapeutic target to decrease this state of low-grade inflammation is the metabolism of the essential amino-acid tryptophan. Its three main metabolic pathways (kynurenine pathway, indole pathway and serotonin/melatonin pathway) result in metabolites such as kynurenic acid, xanturenic acid, indole-3-propionic acid and serotonin/melatonin. The kynurenine pathway is regulated by indoleamine 2,3-dioxygenase (IDO), an enzyme that is upregulated by pro-inflammatory molecules such as INF, IL-6 and LPS. Higher activity of IDO is associated with increased inflammation and fibrosis in NAFLD, as well with increased glucose levels, obesity and atherosclerosis. On the other hand, increased concentrations of the indole pathway metabolites, regulated by the gut microbiome, seem to result in more favorable outcomes. This narrative review summarizes the interactions between tryptophan metabolism, the gut microbiome and the immune system as potential drivers of cardiometabolic diseases in NAFLD.
Collapse
|
30
|
Sabahi S, Homayouni Rad A, Aghebati-Maleki L, Sangtarash N, Ozma MA, Karimi A, Hosseini H, Abbasi A. Postbiotics as the new frontier in food and pharmaceutical research. Crit Rev Food Sci Nutr 2022; 63:8375-8402. [PMID: 35348016 DOI: 10.1080/10408398.2022.2056727] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Food is the essential need of human life and has nutrients that support growth and health. Gastrointestinal tract microbiota involves valuable microorganisms that develop therapeutic effects and are characterized as probiotics. The investigations on appropriate probiotic strains have led to the characterization of specific metabolic byproducts of probiotics named postbiotics. The probiotics must maintain their survival against inappropriate lethal conditions of the processing, storage, distribution, preparation, and digestion system so that they can exhibit their most health effects. Conversely, probiotic metabolites (postbiotics) have successfully overcome these unfavorable conditions and may be an appropriate alternative to probiotics. Due to their specific chemical structure, safe profile, long shelf-life, and the fact that they contain various signaling molecules, postbiotics may have anti-inflammatory, immunomodulatory, antihypertensive properties, inhibiting abnormal cell proliferation and antioxidative activities. Consequently, present scientific literature approves that postbiotics can mimic the fundamental and clinical role of probiotics, and due to their unique characteristics, they can be applied in an oral delivery system (pharmaceutical/functional foods), as a preharvest food safety hurdle, to promote the shelf-life of food products and develop novel functional foods or/and for developing health benefits, and therapeutic aims. This review addresses the latest postbiotic applications with regard to pharmaceutical formulations and commercial food-based products. Potential postbiotic applications in the promotion of host health status, prevention of disease, and complementary treatment are also reviewed.
Collapse
Affiliation(s)
- Sahar Sabahi
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Sangtarash
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahdi Asghari Ozma
- Department of Medical Bacteriology and Virology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Karimi
- Department of Food Safety and Hygiene, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hedayat Hosseini
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Mohamud R, Fawaz M, Kateeb ET, Alkhairy OK, Tayyem R, Lounis M, Al-Raeei M, Dana RK, Al-Ameer HJ, Taha MO, Bindayna KM. Reported Adverse Effects and Attitudes among Arab Populations Following COVID-19 Vaccination: A Large-Scale Multinational Study Implementing Machine Learning Tools in Predicting Post-Vaccination Adverse Effects Based on Predisposing Factors. Vaccines (Basel) 2022; 10:366. [PMID: 35334998 PMCID: PMC8955470 DOI: 10.3390/vaccines10030366] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Background: The unprecedented global spread of coronavirus disease 2019 (COVID-19) has imposed huge challenges on the healthcare facilities, and impacted every aspect of life. This has led to the development of several vaccines against COVID-19 within one year. This study aimed to assess the attitudes and the side effects among Arab communities after receiving a COVID-19 vaccine and use of machine learning (ML) tools to predict post-vaccination side effects based on predisposing factors. Methods: An online-based multinational survey was carried out via social media platforms from 14 June to 31 August 2021, targeting individuals who received at least one dose of a COVID-19 vaccine from 22 Arab countries. Descriptive statistics, correlation, and chi-square tests were used to analyze the data. Moreover, extensive ML tools were utilized to predict 30 post vaccination adverse effects and their severity based on 15 predisposing factors. The importance of distinct predisposing factors in predicting particular side effects was determined using global feature importance employing gradient boost as AutoML. Results: A total of 10,064 participants from 19 Arab countries were included in this study. Around 56% were female and 59% were aged from 20 to 39 years old. A high rate of vaccine hesitancy (51%) was reported among participants. Almost 88% of the participants were vaccinated with one of three COVID-19 vaccines, including Pfizer-BioNTech (52.8%), AstraZeneca (20.7%), and Sinopharm (14.2%). About 72% of participants experienced post-vaccination side effects. This study reports statistically significant associations (p < 0.01) between various predisposing factors and post-vaccinations side effects. In terms of predicting post-vaccination side effects, gradient boost, random forest, and XGBoost outperformed other ML methods. The most important predisposing factors for predicting certain side effects (i.e., tiredness, fever, headache, injection site pain and swelling, myalgia, and sleepiness and laziness) were revealed to be the number of doses, gender, type of vaccine, age, and hesitancy to receive a COVID-19 vaccine. Conclusions: The reported side effects following COVID-19 vaccination among Arab populations are usually non-life-threatening; flu-like symptoms and injection site pain. Certain predisposing factors have greater weight and importance as input data in predicting post-vaccination side effects. Based on the most significant input data, ML can also be used to predict these side effects; people with certain predicted side effects may require additional medical attention, or possibly hospitalization.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia; (M.A.I.A.-H.); (R.M.)
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia; (M.A.I.A.-H.); (R.M.)
| | - Mirna Fawaz
- Nursing Department, Faculty of Health Sciences, Beirut Arab University, Beirut 1105, Lebanon;
| | - Elham T. Kateeb
- Oral Health Research and Promotion Unit, Faculty of Dentistry, Al-Quds University, Jerusalem 51000, Palestine;
| | - Omar K. Alkhairy
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Saudi Arabia;
- King Saud bin Abdulaziz University for Health Sciences, P.O. Box 3660, Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), P.O. Box 3660, Riyadh 11481, Saudi Arabia
| | - Reema Tayyem
- Department of Human Nutrition, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Mohamed Lounis
- Department of Agro-Veterinary Science, Faculty of Natural and Life Sciences, University of Ziane Achour, BP 3117, Djelfa 17000, Algeria;
| | - Marwan Al-Raeei
- Faculty of Sciences, Damascus University, Damascus P.O. Box 30621, Syria;
| | - Rasheed K. Dana
- Faculty of Medicine, Mansoura University, Mansoura, Dakahlia 35516, Egypt;
| | - Hamzeh J. Al-Ameer
- Department of Biology and Biotechnology, Faculty of Science, American University of Madaba, P.O. Box 99, Madaba 17110, Jordan;
| | - Mutasem O. Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Khalid M. Bindayna
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain
| |
Collapse
|
32
|
Xu SS, Zhang XL, Liu SS, Feng ST, Xiang GM, Xu CJ, Fan ZY, Xu K, Wang N, Wang Y, Che JJ, Liu ZG, Mu YL, Li K. Multi-Omic Analysis in a Metabolic Syndrome Porcine Model Implicates Arachidonic Acid Metabolism Disorder as a Risk Factor for Atherosclerosis. Front Nutr 2022; 9:807118. [PMID: 35284467 PMCID: PMC8906569 DOI: 10.3389/fnut.2022.807118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/10/2022] [Indexed: 11/25/2022] Open
Abstract
Background The diet-induced gut microbiota dysbiosis has been suggested as a major risk factor for atherothrombosis, however, the detailed mechanism linking these conditions is yet to be fully understood. Methods We established a long-term excessive-energy diet-induced metabolic syndrome (MetS) inbred Wuzhishan minipig model, which is characterized by its genetic stability, small size, and human-like physiology. The metabolic parameters, atherosclerotic lesions, gut microbiome, and host transcriptome were analyzed. Metabolomics profiling revealed a linkage between gut microbiota and atherothrombosis. Results We showed that white atheromatous plaque was clearly visible on abdominal aorta in the MetS model. Furthermore, using metagenome and metatranscriptome sequencing, we discovered that the long-term excessive energy intake altered the local intestinal microbiota composition and transcriptional profile, which was most dramatically illustrated by the reduced abundance of SCFAs-producing bacteria including Bacteroides, Lachnospiraceae, and Ruminococcaceae in the MetS model. Liver and abdominal aorta transcriptomes in the MetS model indicate that the diet-induced gut microbiota dysbiosis activated host chronic inflammatory responses and significantly upregulated the expression of genes related to arachidonic acid-dependent signaling pathways. Notably, metabolomics profiling further revealed an intimate linkage between arachidonic acid metabolism and atherothrombosis in the host-gut microbial metabolism axis. Conclusions These findings provide new insights into the relationship between atherothrombosis and regulation of gut microbiota via host metabolomes and will be of potential value for the treatment of cardiovascular diseases in MetS.
Collapse
Affiliation(s)
- Song-Song Xu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiu-Ling Zhang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Sha-Sha Liu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Animal Husbandry and Veterinary Department, Beijing Vocational College of Agriculture, Beijing, China
| | - Shu-Tang Feng
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Guang-Ming Xiang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chang-Jiang Xu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zi-Yao Fan
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kui Xu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Nan Wang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yue Wang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jing-Jing Che
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhi-Guo Liu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yu-Lian Mu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Yu-Lian Mu
| | - Kui Li
- State Key Laboratory of Animal Nutrition and Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs of China, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Kui Li
| |
Collapse
|
33
|
Puetz A, Artati A, Adamski J, Schuett K, Romeo F, Stoehr R, Marx N, Federici M, Lehrke M, Kappel BA. Non-targeted metabolomics identify polyamine metabolite acisoga as novel biomarker for reduced left ventricular function. ESC Heart Fail 2022; 9:564-573. [PMID: 34811951 PMCID: PMC8788009 DOI: 10.1002/ehf2.13713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 11/25/2022] Open
Abstract
AIMS Chronic heart failure with reduced ejection fraction remains a major health issue. To date, no reliable biomarker is available to predict reduced left ventricular ejection fraction (LV-EF). We aimed to identify novel circulating biomarkers for reduced left ventricular function using untargeted serum metabolomics in two independent patient cohorts. METHODS AND RESULTS Echocardiography and non-targeted serum metabolomics were conducted in two patient cohorts with varying left ventricular function: (1) 25 patients with type 2 diabetes with established cardiovascular disease or high cardiovascular risk (LV-EF range 20-66%) (discovery cohort) and (2) 37 patients hospitalized for myocardial infarction (LV-EF range 25-60%) (validation cohort). In the discovery cohort, untargeted metabolomics revealed seven metabolites performing better than N-terminal pro-B-type natriuretic peptide in the prediction of impaired left ventricular function shown by LV-EF. For only one of the metabolites, acisoga, the predictive value for LV-EF could be confirmed in the validation cohort (r = -0.37, P = 0.02). In the discovery cohort, acisoga did not only correlate with LV-EF (r = -60, P = 0.0016), but also with global circumferential strain (r = 0.67, P = 0.0003) and global longitudinal strain (r = 0.68, P = 0.0002). Similar results could be detected in the discovery cohort in a 6 month follow-up proofing stability of these results over time. With an area under the curve of 0.86 in the receiver operating characteristic analysis, acisoga discriminated between patients with normal EF and LV-EF < 40%. Multivariate analysis exposed acisoga as independent marker for impairment of LV-EF (Beta = -0.71, P = 0.004). CONCLUSIONS We found the polyamine metabolite acisoga to be elevated in patients with impaired LV-EF in two independent cohorts. Our analyses suggest that acisoga may be a valuable biomarker to detect patients with heart failure with reduced ejection fraction.
Collapse
Affiliation(s)
- Andreas Puetz
- Department of Internal Medicine 1, University Hospital AachenRWTH Aachen UniversityAachenGermany
| | - Anna Artati
- Metabolomics and Proteomics CoreHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Jerzy Adamski
- Institute of Experimental GeneticsHelmholtz Zentrum München, German Research Center for Environmental HealthNeuherbergGermany
- Institute of Biochemistry, Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
- Department of Biochemistry, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Katharina Schuett
- Department of Internal Medicine 1, University Hospital AachenRWTH Aachen UniversityAachenGermany
| | - Francesco Romeo
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
| | - Robert Stoehr
- Department of Internal Medicine 1, University Hospital AachenRWTH Aachen UniversityAachenGermany
| | - Nikolaus Marx
- Department of Internal Medicine 1, University Hospital AachenRWTH Aachen UniversityAachenGermany
| | - Massimo Federici
- Department of Systems MedicineUniversity of Rome Tor VergataRomeItaly
- Center for AtherosclerosisPoliclinico Tor VergataRomeItaly
| | - Michael Lehrke
- Department of Internal Medicine 1, University Hospital AachenRWTH Aachen UniversityAachenGermany
| | - Ben A. Kappel
- Department of Internal Medicine 1, University Hospital AachenRWTH Aachen UniversityAachenGermany
| |
Collapse
|
34
|
Gut microbiota and its role in stress-induced hyperalgesia: gender-specific responses linked to different changes in serum metabolites. Pharmacol Res 2022; 177:106129. [DOI: 10.1016/j.phrs.2022.106129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
|
35
|
Garcia-Irigoyen O, Bovenga F, Piglionica M, Piccinin E, Cariello M, Arconzo M, Peres C, Corsetto PA, Rizzo AM, Ballanti M, Menghini R, Mingrone G, Lefebvre P, Staels B, Shirasawa T, Sabbà C, Villani G, Federici M, Moschetta A. Enterocyte superoxide dismutase 2 deletion drives obesity. iScience 2022; 25:103707. [PMID: 35036884 PMCID: PMC8753186 DOI: 10.1016/j.isci.2021.103707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/19/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
Compelling evidence support an involvement of oxidative stress and intestinal inflammation as early events in the predisposition and development of obesity and its related comorbidities. Here, we show that deficiency of the major mitochondrial antioxidant enzyme superoxide dismutase 2 (SOD2) in the gastrointestinal tract drives spontaneous obesity. Intestinal epithelium-specific Sod2 ablation in mice induced adiposity and inflammation via phospholipase A2 (PLA2) activation and increased release of omega-6 polyunsaturated fatty acid arachidonic acid. Remarkably, this obese phenotype was rescued when fed an essential fatty acid-deficient diet, which abrogates de novo biosynthesis of arachidonic acid. Data from clinical samples revealed that the negative correlation between intestinal Sod2 mRNA levels and obesity features appears to be conserved between mice and humans. Collectively, our findings suggest a role of intestinal Sod2 levels, PLA2 activity, and arachidonic acid in obesity presenting new potential targets of therapeutic interest in the context of this metabolic disorder.
Collapse
Affiliation(s)
- Oihane Garcia-Irigoyen
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Fabiola Bovenga
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marilidia Piglionica
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Elena Piccinin
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy.,Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marica Cariello
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Maria Arconzo
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Claudia Peres
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Paola Antonia Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via D. Trentacoste 2, 20133 Milan, Italy
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via D. Trentacoste 2, 20133 Milan, Italy
| | - Marta Ballanti
- Center for Atherosclerosis, Policlinico Tor Vergata, 00133 Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Diabetes and Nutritional Sciences, Hodgkin Building, Guy's Campus, King's College London, London, UK
| | - Philippe Lefebvre
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Takuji Shirasawa
- Department of Molecular Gerontology, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Carlo Sabbà
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Gaetano Villani
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Massimo Federici
- Center for Atherosclerosis, Policlinico Tor Vergata, 00133 Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonio Moschetta
- Clinica Medica "Cesare Frugoni", Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124 Bari, Italy.,IRCCS Istituto Tumori "Giovanni Paolo II", Viale O. Flacco 65, 70124 Bari, Italy
| |
Collapse
|
36
|
Yoo JY, Sniffen S, McGill Percy KC, Pallaval VB, Chidipi B. Gut Dysbiosis and Immune System in Atherosclerotic Cardiovascular Disease (ACVD). Microorganisms 2022; 10:108. [PMID: 35056557 PMCID: PMC8780459 DOI: 10.3390/microorganisms10010108] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular disease and mortality worldwide. Alterations in the gut microbiota composition, known as gut dysbiosis, have been shown to contribute to atherosclerotic cardiovascular disease (ACVD) development through several pathways. Disruptions in gut homeostasis are associated with activation of immune processes and systemic inflammation. The gut microbiota produces several metabolic products, such as trimethylamine (TMA), which is used to produce the proatherogenic metabolite trimethylamine-N-oxide (TMAO). Short-chain fatty acids (SCFAs), including acetate, butyrate, and propionate, and certain bile acids (BAs) produced by the gut microbiota lead to inflammation resolution and decrease atherogenesis. Chronic low-grade inflammation is associated with common risk factors for atherosclerosis, including metabolic syndrome, type 2 diabetes mellitus (T2DM), and obesity. Novel strategies for reducing ACVD include the use of nutraceuticals such as resveratrol, modification of glucagon-like peptide 1 (GLP-1) levels, supplementation with probiotics, and administration of prebiotic SCFAs and BAs. Investigation into the relationship between the gut microbiota, and its metabolites, and the host immune system could reveal promising insights into ACVD development, prognostic factors, and treatments.
Collapse
Affiliation(s)
- Ji Youn Yoo
- College of Nursing, University of Tennessee, 1200 Volunteer Blvd, Knoxville, TN 37996, USA
| | - Sarah Sniffen
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kyle Craig McGill Percy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Bojjibabu Chidipi
- Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 78, Tampa, FL 33612, USA
| |
Collapse
|
37
|
Liu D, Wang J, Zeng H, Zhou F, Wen B, Zhang X, Luo Y, Wu W, Huang J, Liu Z. The metabolic regulation of Fuzhuan brick tea in high-fat diet-induced obese mice and the potential contribution of gut microbiota. Food Funct 2022; 13:356-374. [PMID: 34904994 DOI: 10.1039/d1fo02181h] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This study investigated the metabolic effects of Fuzhuan brick tea (FBT) in high-fat diet (HFD)-induced obese mice and the potential contribution of gut microbiota. The results showed that FBT ameliorated the HFD-induced glycerophospholipid metabolic aberrance, specifically increased the serum levels of phosphatidylcholines (PCs), lysophosphatidylcholines (LysoPCs), and the ratio of PC to phosphatidylethanolamines (PE). Besides, FBT increased the serum level of gut microbiota-derived aryl hydrocarbon receptor (AhR) ligand, 3-indole propionic acid, as well as the relative abundance of intestinal AhR-ligand producing bacteria such as Clostridiaceae, Bacteroidales_S24-7_group, and Lactobacillaceae. However, the metabolic benefits of FBT were weakened when the gut microbiota were depleted by antibiotic treatment, thereby suggesting that gut microbiota was required for FBT to regulate glycerophospholipid metabolism. Indeed, the metabolites regulated by FBT were significantly correlated with the AhR-ligand producing bacteria. The KEGG pathway enrichment analysis and expressions of AhR target genes indicated that FBT would improve the glycerophospholipid metabolism via the AhR-Pemt signal axis, in which the gut microbiota and their metabolites played pivotal mediators. Overall, FBT could be a functional beverage to improve HFD-induced metabolic disorders in a gut microbiota dependent manner.
Collapse
Affiliation(s)
- Dongmin Liu
- Changsha University of Science & Technology, Changsha 410114, China.,Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Jianhui Wang
- Changsha University of Science & Technology, Changsha 410114, China
| | - Hongliang Zeng
- Research Institute of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, China
| | - Fang Zhou
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Beibei Wen
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Xiangna Zhang
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Yong Luo
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Wenliang Wu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Jianan Huang
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Zhonghua Liu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| |
Collapse
|
38
|
Alam MJ, Puppala V, Uppulapu SK, Das B, Banerjee SK. Human microbiome and cardiovascular diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:231-279. [PMID: 36280321 DOI: 10.1016/bs.pmbts.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Mayneris-Perxachs J, Moreno-Navarrete JM, Ballanti M, Monteleone G, Alessandro Paoluzi O, Mingrone G, Lefebvre P, Staels B, Federici M, Puig J, Garre J, Ramos R, Fernández-Real JM. Lipidomics and metabolomics signatures of SARS-CoV-2 mediators/receptors in peripheral leukocytes, jejunum and colon. Comput Struct Biotechnol J 2021; 19:6080-6089. [PMID: 34777716 PMCID: PMC8574068 DOI: 10.1016/j.csbj.2021.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 12/14/2022] Open
Abstract
Cell surface receptor-mediated viral entry plays a critical role in this infection. Well-established SARS-CoV-2 receptors such as ACE2 and TMPRSS2 are highly expressed in the gastrointestinal tract. In fact, there are evidences that SARS-CoV-2 infects epithelial cells from the digestive system. However, emerging research has identified novel mediators such as DPP9, TYK2, and CCR2, all playing a critical role in inflammation. We evaluated the expression of SARS-CoV-2 receptors in peripheral leukocytes (n = 469), jejunum (n = 30), and colon (n = 37) of three independent cohorts by real-time PCR, RNA-sequencing, and microarray transcriptomics. We also performed HPCL-MS/MS lipidomics and metabolomics analyses to identify signatures linked to SARS-CoV-2 receptors. We found markedly higher peripheral leukocytes ACE2 expression levels in women compared to men, whereas the intestinal expression of TMPRSS2 was positively associated with BMI. Consistent lipidomics signatures associated with the expression of these mediators were found in both tissues and peripheral leukocytes involving n-3 long-chain PUFAs and arachidonic acid-derived eicosanoids, which play a key role in the regulation of inflammation and may interfere with viral entry and replication. Medium- and long-chain hydroxy acids, which have shown to interfere in viral replication, were also liked to SARS-CoV2 receptors. Gonadal steroids were also associated with the expression of some of these receptors, even after controlling for sex. The expression of SARS-CoV2 receptors was associated with several metabolic and nutritional traits in different cell types. This information may be useful in the design of potential therapies targeted at coronavirus entry.
Collapse
Affiliation(s)
- Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain.,Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.,Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Catalonia, Spain
| | - José Maria Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain.,Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.,Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Catalonia, Spain
| | - Marta Ballanti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy
| | | | | | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Diabetes and Nutritional Sciences, Hodgkin Building, Guy's Campus, King's College London, London, United Kingdom
| | - Philippe Lefebvre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy
| | - Josep Puig
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Catalonia, Spain.,Institute of Diagnostic Imaging (IDI)-Research Unit (IDIR), Parc Sanitari Pere Virgili, Barcelona, Spain.,Medical Imaging, Girona Biomedical Research Institute (IdibGi), Girona, Spain.,Department of Radiology (IDI), Dr. Josep Trueta University Hospital, Girona, Spain
| | - Josep Garre
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Catalonia, Spain.,Research Group on Aging, Disability and Health, Girona Biomedical Research Institute (IdIBGi), Girona, Spain.,Serra-Húnter Professor. Department of Nursing, University of Girona, Girona Spain
| | - Rafael Ramos
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Catalonia, Spain.,Vascular Health Research Group of Girona (ISV-Girona). Jordi Gol Institute for Primary Care Research (Institut Universitari per a la RecercaenAtencióPrimària Jordi Gol I Gorina -IDIAPJGol), Catalonia, Spain
| | - José-Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain.,Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain.,Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.,Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Catalonia, Spain
| |
Collapse
|
40
|
Hatmal MM, Alshaer W, Mahmoud IS, Al-Hatamleh MAI, Al-Ameer HJ, Abuyaman O, Zihlif M, Mohamud R, Darras M, Al Shhab M, Abu-Raideh R, Ismail H, Al-Hamadi A, Abdelhay A. Investigating the association of CD36 gene polymorphisms (rs1761667 and rs1527483) with T2DM and dyslipidemia: Statistical analysis, machine learning based prediction, and meta-analysis. PLoS One 2021; 16:e0257857. [PMID: 34648514 PMCID: PMC8516279 DOI: 10.1371/journal.pone.0257857] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/11/2021] [Indexed: 12/15/2022] Open
Abstract
CD36 (cluster of differentiation 36) is a membrane protein involved in lipid metabolism and has been linked to pathological conditions associated with metabolic disorders, such as diabetes and dyslipidemia. A case-control study was conducted and included 177 patients with type-2 diabetes mellitus (T2DM) and 173 control subjects to study the involvement of CD36 gene rs1761667 (G>A) and rs1527483 (C>T) polymorphisms in the pathogenesis of T2DM and dyslipidemia among Jordanian population. Lipid profile, blood sugar, gender and age were measured and recorded. Also, genotyping analysis for both polymorphisms was performed. Following statistical analysis, 10 different neural networks and machine learning (ML) tools were used to predict subjects with diabetes or dyslipidemia. Towards further understanding of the role of CD36 protein and gene in T2DM and dyslipidemia, a protein-protein interaction network and meta-analysis were carried out. For both polymorphisms, the genotypic frequencies were not significantly different between the two groups (p > 0.05). On the other hand, some ML tools like multilayer perceptron gave high prediction accuracy (≥ 0.75) and Cohen's kappa (κ) (≥ 0.5). Interestingly, in K-star tool, the accuracy and Cohen's κ values were enhanced by including the genotyping results as inputs (0.73 and 0.46, respectively, compared to 0.67 and 0.34 without including them). This study confirmed, for the first time, that there is no association between CD36 polymorphisms and T2DM or dyslipidemia among Jordanian population. Prediction of T2DM and dyslipidemia, using these extensive ML tools and based on such input data, is a promising approach for developing diagnostic and prognostic prediction models for a wide spectrum of diseases, especially based on large medical databases.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
- * E-mail:
| | - Walhan Alshaer
- Cell Therapy Centre, The University of Jordan, Amman, Jordan
| | - Ismail S. Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Hamzeh J. Al-Ameer
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Omar Abuyaman
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Mais Darras
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Mohammad Al Shhab
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Rand Abu-Raideh
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Hilweh Ismail
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Ali Al-Hamadi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Ali Abdelhay
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
41
|
Alhajri N, Khursheed R, Ali MT, Abu Izneid T, Al-Kabbani O, Al-Haidar MB, Al-Hemeiri F, Alhashmi M, Pottoo FH. Cardiovascular Health and The Intestinal Microbial Ecosystem: The Impact of Cardiovascular Therapies on The Gut Microbiota. Microorganisms 2021; 9:2013. [PMID: 34683334 PMCID: PMC8541580 DOI: 10.3390/microorganisms9102013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023] Open
Abstract
It has become evident over the past several years that the intestinal microbial ecosystem plays a critical role in the development and prevention of cardiovascular diseases (CVDs) and other metabolic disorders, such as hypertension, obesity, diabetes mellitus, and metabolic syndrome. The intestinal microbiota ecosystem functions as a major virtual endocrine organ that interacts and responds to molecules' signals within the host. Several meta-organismal pathways are involved in the gut-host interaction, including trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFA). Host phenotype and cardiovascular diseases (CVDs) varying from hypertension, insulin resistance, and obesity to more specific inflammatory processes, such as atherosclerosis and hypercoagulability, have shown to be affected by the gut-host interaction. Additionally, several studies that involved animals and humans demonstrated a striking connection between the development of new CVDs and an imbalance in the gut microbiota composition along with the presence of their derived metabolites. Through this review article, we aim to evaluate the role of the normal gut microbiota ecosystem, its association with CVDs, effects of the therapies used to control and manage CVDs in the gut microbiota environment and explore potential therapeutic interventions to amplify disease outcomes in patients with CVDs.
Collapse
Affiliation(s)
- Noora Alhajri
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
- Department of Medicine, Sheikh Shakhbout Medical City (SSMC), Abu Dhabi P.O. Box 11001, United Arab Emirates
| | - Rubiya Khursheed
- Department of Pharmaceutical Sciences, Lovely Professional University, Punjab 144403, India;
| | - Mohammad Taher Ali
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia; (M.T.A.); (F.H.P.)
| | - Tareq Abu Izneid
- Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, Abu Dhabi P.O. Box 112612, United Arab Emirates;
| | - Oumaima Al-Kabbani
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Mahdia B. Al-Haidar
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Fatima Al-Hemeiri
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Mohamed Alhashmi
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (O.A.-K.); (M.B.A.-H.); (F.A.-H.); (M.A.)
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia; (M.T.A.); (F.H.P.)
| |
Collapse
|
42
|
Su J, Zhao Q, Zhao A, Jia W, Zhu W, Lu J, Ma X. Serum metabolic signatures of subclinical atherosclerosis in patients with type 2 diabetes mellitus: a preliminary study. Acta Diabetol 2021; 58:1217-1224. [PMID: 33871690 DOI: 10.1007/s00592-021-01717-7] [Citation(s) in RCA: 4] [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: 01/01/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022]
Abstract
AIMS Atherosclerotic cardiovascular disease remains the leading cause of death among patients with diabetes. Early identification of subclinical atherosclerosis is essential for the management of diabetic patients. This study aimed to characterize serum metabolic signatures associated with carotid intima-media thickness (C-IMT), a proxy of subclinical atherosclerosis, in patients with type 2 diabetes mellitus (T2DM). METHODS After 1:1 matching by sex, age, body mass index, glycated haemoglobin A1c, and other clinical parameters, a total of 462 T2DM patients were enrolled, consisting of 231 patients with C-IMT of ≥ 1 mm (abnormal C-IMT) and 231 patients with C-IMT of < 1 mm (normal C-IMT). C-IMT was assessed using ultrasonography. The serum metabolic profiling of fasting blood samples was performed using liquid chromatography-tandem triple quadrupole mass spectrometer coupled with the multivariate and univariate statistical analysis. RESULTS Patients with abnormal C-IMT had significantly higher deoxycholic acid (DCA) and taurodeoxycholic acid (TDCA) levels, and lower levels of taurocholic acid (TCA) than those with normal C-IMT. Conditional logistic regression analysis revealed that per 1-standard deviation increase of DCA, TDCA and TCA were significantly associated with 64.7% (95% CI: 1.234-2.196) and 38.5% (95% CI: 1.124-1.706) higher, and 26.8% (95% CI: 0.597-0.897) lower risk of abnormal C-IMT, after adjustment of confounders. The addition of DCA, TCA, or DCA × TDCA/TCA ratio significantly improved the discrimination of abnormal C-IMT over traditional risk factors. CONCLUSIONS Serum bile acids may be potential biomarkers for subclinical atherosclerosis in T2DM patients, which needs further confirmation.
Collapse
Affiliation(s)
- Jiaorong Su
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, 600 Yishan Road, Shanghai, 200233, China
| | - Qing Zhao
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Aihua Zhao
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Wei Jia
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Wei Zhu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, 600 Yishan Road, Shanghai, 200233, China
| | - Jingyi Lu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, 600 Yishan Road, Shanghai, 200233, China.
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
43
|
Kumar P, Lee JH, Lee J. Diverse roles of microbial indole compounds in eukaryotic systems. Biol Rev Camb Philos Soc 2021; 96:2522-2545. [PMID: 34137156 PMCID: PMC9290978 DOI: 10.1111/brv.12765] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Indole and its derivatives are widespread across different life forms, functioning as signalling molecules in prokaryotes and with more diverse roles in eukaryotes. A majority of indoles found in the environment are attributed to bacterial enzymes converting tryptophan into indole and its derivatives. The involvement of indoles among lower organisms as an interspecies and intraspecies signal is well known, with many reports showing that inter‐kingdom interactions involving microbial indole compounds are equally important as they influence defence systems and even the behaviour of higher organisms. This review summarizes recent advances in our understanding of the functional properties of indole and indole derivatives in diverse eukaryotes. Furthermore, we discuss current perspectives on the role of microbial indoles in human diseases such as diabetes, obesity, atherosclerosis, and cancers. Deciphering the function of indoles as biomarkers of metabolic state will facilitate the formulation of diet‐based treatments and open unique therapeutic opportunities.
Collapse
Affiliation(s)
- Prasun Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| |
Collapse
|
44
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Hatmal M, Alhaj-Qasem DM, Olaimat TM, Mohamud R. Side Effects and Perceptions Following COVID-19 Vaccination in Jordan: A Randomized, Cross-Sectional Study Implementing Machine Learning for Predicting Severity of Side Effects. Vaccines (Basel) 2021; 9:vaccines9060556. [PMID: 34073382 PMCID: PMC8229440 DOI: 10.3390/vaccines9060556] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Since the coronavirus disease 2019 (COVID-19) was declared a pandemic, there was no doubt that vaccination is the ideal protocol to tackle it. Within a year, a few COVID-19 vaccines have been developed and authorized. This unparalleled initiative in developing vaccines created many uncertainties looming around the efficacy and safety of these vaccines. This study aimed to assess the side effects and perceptions following COVID-19 vaccination in Jordan. Methods: A cross-sectional study was conducted by distributing an online survey targeted toward Jordan inhabitants who received any COVID-19 vaccines. Data were statistically analyzed and certain machine learning (ML) tools, including multilayer perceptron (MLP), eXtreme gradient boosting (XGBoost), random forest (RF), and K-star were used to predict the severity of side effects. Results: A total of 2213 participants were involved in the study after receiving Sinopharm, AstraZeneca, Pfizer-BioNTech, and other vaccines (38.2%, 31%, 27.3%, and 3.5%, respectively). Generally, most of the post-vaccination side effects were common and non-life-threatening (e.g., fatigue, chills, dizziness, fever, headache, joint pain, and myalgia). Only 10% of participants suffered from severe side effects; while 39% and 21% of participants had moderate and mild side effects, respectively. Despite the substantial variations between these vaccines in the presence and severity of side effects, the statistical analysis indicated that these vaccines might provide the same protection against COVID-19 infection. Finally, around 52.9% of participants suffered before vaccination from vaccine hesitancy and anxiety; while after vaccination, 95.5% of participants have advised others to get vaccinated, 80% felt more reassured, and 67% believed that COVID-19 vaccines are safe in the long term. Furthermore, based on the type of vaccine, demographic data, and side effects, the RF, XGBoost, and MLP gave both high accuracies (0.80, 0.79, and 0.70, respectively) and Cohen’s kappa values (0.71, 0.70, and 0.56, respectively). Conclusions: The present study confirmed that the authorized COVID-19 vaccines are safe and getting vaccinated makes people more reassured. Most of the post-vaccination side effects are mild to moderate, which are signs that body’s immune system is building protection. ML can also be used to predict the severity of side effects based on the input data; predicted severe cases may require more medical attention or even hospitalization.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan
- Correspondence: (M.M.H.); (R.M.)
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa 13133, Jordan;
| | | | | | | | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
- Correspondence: (M.M.H.); (R.M.)
| |
Collapse
|
45
|
Murovec B, Deutsch L, Stres B. General Unified Microbiome Profiling Pipeline (GUMPP) for Large Scale, Streamlined and Reproducible Analysis of Bacterial 16S rRNA Data to Predicted Microbial Metagenomes, Enzymatic Reactions and Metabolic Pathways. Metabolites 2021; 11:336. [PMID: 34074026 PMCID: PMC8225202 DOI: 10.3390/metabo11060336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/14/2021] [Accepted: 05/23/2021] [Indexed: 11/23/2022] Open
Abstract
General Unified Microbiome Profiling Pipeline (GUMPP) was developed for large scale, streamlined and reproducible analysis of bacterial 16S rRNA data and prediction of microbial metagenomes, enzymatic reactions and metabolic pathways from amplicon data. GUMPP workflow introduces reproducible data analyses at each of the three levels of resolution (genus; operational taxonomic units (OTUs); amplicon sequence variants (ASVs)). The ability to support reproducible analyses enables production of datasets that ultimately identify the biochemical pathways characteristic of disease pathology. These datasets coupled to biostatistics and mathematical approaches of machine learning can play a significant role in extraction of truly significant and meaningful information from a wide set of 16S rRNA datasets. The adoption of GUMPP in the gut-microbiota related research enables focusing on the generation of novel biomarkers that can lead to the development of mechanistic hypotheses applicable to the development of novel therapies in personalized medicine.
Collapse
Affiliation(s)
- Boštjan Murovec
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, SI-1000 Ljubljana, Slovenia;
| | - Leon Deutsch
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia;
| | - Blaž Stres
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia;
- Faculty of Civil and Geodetic Engineering, University of Ljubljana, Jamova 2, SI-1000 Ljubljana, Slovenia
- Department of Automation, Jožef Stefan Institute, Biocybernetics and Robotics, Jamova 39, SI-1000 Ljubljana, Slovenia
- Department of Microbiology, University of Innsbruck, Technikerstrasse 25d, A-6020 Innsbruck, Austria
| |
Collapse
|
46
|
Garshick MS, Nikain C, Tawil M, Pena S, Barrett TJ, Wu BG, Gao Z, Blaser MJ, Fisher EA. Reshaping of the gastrointestinal microbiome alters atherosclerotic plaque inflammation resolution in mice. Sci Rep 2021; 11:8966. [PMID: 33903700 PMCID: PMC8076321 DOI: 10.1038/s41598-021-88479-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Since alterations in the intestinal microbiota may induce systemic inflammation and polarization of macrophages to the M1 state, the microbiome role in atherosclerosis, an M1-driven disease, requires evaluation. We aimed to determine if antibiotic (Abx) induced alterations to the intestinal microbiota interferes with atherosclerotic plaque inflammation resolution after lipid-lowering in mice. Hyperlipidemic Apoe−/− mice were fed a western diet to develop aortic atherosclerosis with aortas then transplanted into normolipidemic wild-type (WT) mice to model clinically aggressive lipid management and promote atherosclerosis inflammation resolution. Gut microbial composition pre and post-transplant was altered via an enteral antibiotic or not. Post aortic transplant, after Abx treatment, while plaque size did not differ, compared to Apoe−/− mice, Abx– WT recipient mice had a 32% reduction in CD68-expressing cells (p = 0.02) vs. a non-significant 12% reduction in Abx+ WT mice. A trend toward an M1 plaque CD68-expresing cell phenotype was noted in Abx+ mice. By 16S rRNA sequence analysis, the Abx+ mice had reduced alpha diversity and increased Firmicutes/Bacteroidetes relative abundance ratio with a correlation between gut Firmicutes abundance and plaque CD68-expressing cell content (p < 0.05). These results indicate that in a murine atherosclerotic plaque inflammation resolution model, antibiotic-induced microbiome perturbation may blunt the effectiveness of lipid-lowering to reduce the content of plaque inflammatory CD68-expressing cells.
Collapse
Affiliation(s)
- Michael S Garshick
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine, New York, USA.,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Cyrus Nikain
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Michael Tawil
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Stephanie Pena
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Tessa J Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA
| | - Benjamin G Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University School of Medicine, New York, USA.,Division of Pulmonary and Critical Care, Veterans Affairs New York Harbor Healthcare System, New York, NY, USA
| | - Zhan Gao
- Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane West, Piscataway, NJ, 08854, USA.
| | - Edward A Fisher
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine, New York, USA. .,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, USA. .,Marc and Ruti Bell Vascular Biology Program, Cardiovascular Research Center, New York University Langone Health, New York, USA.
| |
Collapse
|
47
|
Gesper M, Nonnast ABH, Kumowski N, Stoehr R, Schuett K, Marx N, Kappel BA. Gut-Derived Metabolite Indole-3-Propionic Acid Modulates Mitochondrial Function in Cardiomyocytes and Alters Cardiac Function. Front Med (Lausanne) 2021; 8:648259. [PMID: 33829028 PMCID: PMC8019752 DOI: 10.3389/fmed.2021.648259] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background: The gut microbiome has been linked to the onset of cardiometabolic diseases, in part facilitated through gut microbiota-dependent metabolites such as trimethylamine-N-oxide. However, molecular pathways associated to heart failure mediated by microbial metabolites remain largely elusive. Mitochondria play a pivotal role in cellular energy metabolism and mitochondrial dysfunction has been associated to heart failure pathogenesis. Aim of the current study was to evaluate the impact of gut-derived metabolites on mitochondrial function in cardiomyocytes via an in vitro screening approach. Methods: Based on a systematic Medline research, 25 microbial metabolites were identified and screened for their metabolic impact with a focus on mitochondrial respiration in HL-1 cardiomyocytes. Oxygen consumption rate in response to different modulators of the respiratory chain were measured by a live-cell metabolic assay platform. For one of the identified metabolites, indole-3-propionic acid, studies on specific mitochondrial complexes, cytochrome c, fatty acid oxidation, mitochondrial membrane potential, and reactive oxygen species production were performed. Mitochondrial function in response to this metabolite was further tested in human hepatic and endothelial cells. Additionally, the effect of indole-3-propionic acid on cardiac function was studied in isolated perfused hearts of C57BL/6J mice. Results: Among the metabolites examined, microbial tryptophan derivative indole-3-propionic acid could be identified as a modulator of mitochondrial function in cardiomyocytes. While acute treatment induced enhancement of maximal mitochondrial respiration (+21.5 ± 7.8%, p < 0.05), chronic exposure led to mitochondrial dysfunction (-18.9 ± 9.1%; p < 0.001) in cardiomyocytes. The latter effect of indole-3-propionic acids could also be observed in human hepatic and endothelial cells. In isolated perfused mouse hearts, indole-3-propionic acid was dose-dependently able to improve cardiac contractility from +26.8 ± 11.6% (p < 0.05) at 1 μM up to +93.6 ± 14.4% (p < 0.001) at 100 μM. Our mechanistic studies on indole-3-propionic acids suggest potential involvement of fatty acid oxidation in HL-1 cardiomyocytes. Conclusion: Our data indicate a direct impact of microbial metabolites on cardiac physiology. Gut-derived metabolite indole-3-propionic acid was identified as mitochondrial modulator in cardiomyocytes and altered cardiac function in an ex vivo mouse model.
Collapse
Affiliation(s)
- Maren Gesper
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Alena B H Nonnast
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nina Kumowski
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Robert Stoehr
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Katharina Schuett
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nikolaus Marx
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ben A Kappel
- Department of Internal Medicine 1, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
48
|
Microbiota-Mediated Immune Regulation in Atherosclerosis. Molecules 2021; 26:molecules26010179. [PMID: 33401401 PMCID: PMC7795654 DOI: 10.3390/molecules26010179] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
There is a high level of interest in identifying metabolites of endogenously produced or dietary compounds generated by the gastrointestinal (GI) tract microbiota, and determining the functions of these metabolites in health and disease. There is a wealth of compelling evidence that the microbiota is linked with many complex chronic inflammatory diseases, including atherosclerosis. Macrophages are key target immune cells in atherosclerosis. A hallmark of atherosclerosis is the accumulation of pro-inflammatory macrophages in coronary arteries that respond to pro-atherogenic stimuli and failure of digesting lipids that contribute to foam cell formation in atherosclerotic plaques. This review illustrates the role of tryptophan-derived microbiota metabolites as an aryl hydrocarbon receptor (AhR) ligand that has immunomodulatory properties. Also, microbiota-dependent trimethylamine-N-oxide (TMAO) metabolite production is associated with a deleterious effect that promotes atherosclerosis, and metabolite indoxyl sulfate has been shown to exacerbate atherosclerosis. Our objective in this review is to discuss the role of microbiota-derived metabolites in atherosclerosis, specifically the consequences of microbiota-induced effects of innate immunity in response to atherogenic stimuli, and how specific beneficial/detrimental metabolites impact the development of atherosclerosis by regulating chronic endotoxemic and lipotoxic inflammation.
Collapse
|
49
|
Effects of Antibiotics upon the Gut Microbiome: A Review of the Literature. Biomedicines 2020; 8:biomedicines8110502. [PMID: 33207631 PMCID: PMC7696078 DOI: 10.3390/biomedicines8110502] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
The human gastrointestinal tract carries a large number of microorganisms associated with complex metabolic processes and interactions. Although antibiotic treatment is crucial for combating infections, its negative effects on the intestinal microbiota and host immunity have been shown to be of the utmost importance. Multiple studies have recognized the adverse consequences of antibiotic use upon the gut microbiome in adults and neonates, causing dysbiosis of the microbiota. Repeated antibiotic treatments in clinical care or low-dosage intake from food could be contributing factors in this issue. Researchers in both human and animal studies have strived to explain this multifaceted relationship. The present review intends to elucidate the axis of the gastrointestinal microbiota and antibiotics resistance and to highlight the main aspects of the issue.
Collapse
|
50
|
Kiouptsi K, Pontarollo G, Todorov H, Braun J, Jäckel S, Koeck T, Bayer F, Karwot C, Karpi A, Gerber S, Jansen Y, Wild P, Ruf W, Daiber A, Van Der Vorst E, Weber C, Döring Y, Reinhardt C. Germ-free housing conditions do not affect aortic root and aortic arch lesion size of late atherosclerotic low-density lipoprotein receptor-deficient mice. Gut Microbes 2020; 11:1809-1823. [PMID: 32579470 PMCID: PMC7524356 DOI: 10.1080/19490976.2020.1767463] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The microbiota has been linked to the development of atherosclerosis, but the functional impact of these resident bacteria on the lesion size and cellular composition of atherosclerotic plaques in the aorta has never been experimentally addressed with the germ-free low-density lipoprotein receptor-deficient (Ldlr-/- ) mouse atherosclerosis model. Here, we report that 16 weeks of high-fat diet (HFD) feeding of hypercholesterolemic Ldlr-/- mice at germ-free (GF) housing conditions did not impact relative aortic root plaque size, macrophage content, and necrotic core area. Likewise, we did not find changes in the relative aortic arch lesion size. However, late atherosclerotic GF Ldlr-/- mice had altered inflammatory plasma protein markers and reduced smooth muscle cell content in their atherosclerotic root plaques relative to CONV-R Ldlr-/- mice. Neither absolute nor relative aortic root or aortic arch plaque size correlated with age. Our analyses on GF Ldlr-/- mice did not reveal a significant contribution of the microbiota in late aortic atherosclerosis.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Giulia Pontarollo
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Hristo Todorov
- Institute of Developmental Biology and Neurobiology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Johannes Braun
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Sven Jäckel
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany,German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany
| | - Thomas Koeck
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany,German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Franziska Bayer
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Cornelia Karwot
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Angelica Karpi
- Center for Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Susanne Gerber
- Institute of Developmental Biology and Neurobiology, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Yvonne Jansen
- Institute of Cardiovascular Prevention, Department of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Philipp Wild
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany,German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany,German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany,Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, USA
| | - Andreas Daiber
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany,Center for Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany
| | - Emiel Van Der Vorst
- Institute of Cardiovascular Prevention, Department of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands,Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Christian Weber
- Institute of Cardiovascular Prevention, Department of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Yvonne Döring
- Institute of Cardiovascular Prevention, Department of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany,Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany,German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany,CONTACT Christoph Reinhardt University Medical Center Mainz, Mainz55131, Germany
| |
Collapse
|