1
|
Tong Y, Guo S, Li T, Yang K, Gao W, Peng F, Zou X. Gut microbiota and renal fibrosis. Life Sci 2024; 357:123072. [PMID: 39307181 DOI: 10.1016/j.lfs.2024.123072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Renal fibrosis represents a critical pathological condition in the progression of renal dysfunction, characterized by aberrant accumulation of extracellular matrix (ECM) and structural alterations in renal tissue. Recent research has highlighted the potential significance of gut microbiota and demonstrated their influence on host health and disease mechanisms through the production of bioactive metabolites. This review examines the role of alterations in gut microbial composition and their metabolites in the pathophysiological processes underlying renal fibrosis. It delineates current therapeutic interventions aimed at modulating gut microbiota composition, encompassing dietary modifications, pharmacological approaches, and probiotic supplementation, while evaluating their efficacy in mitigating renal fibrosis. Through a comprehensive analysis of current research findings, this review enhances our understanding of the bidirectional interaction between gut microbiota and renal fibrosis, establishing a theoretical foundation for future research directions and potential clinical applications in this domain.
Collapse
Affiliation(s)
- Yinghao Tong
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Shangze Guo
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Ting Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Kexin Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Fujun Peng
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
2
|
Zhang WW, Huo JL, Xiao MD, Xu YJ, Zhou J. Exploring the potential link between gut microbiota and chronic kidney disease in causality: A 2-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40236. [PMID: 39470494 PMCID: PMC11521073 DOI: 10.1097/md.0000000000040236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024] Open
Abstract
Increasing evidence indicates a significant correlation between gut microbiota (GM) and susceptibility to chronic kidney disease (CKD). However, causal relationship presence remains uncertain. Mendelian randomization (MR) was applied to evaluate potential causal relation from GM to CKD. Genomic association analysis aggregates publicly online databases, utilizing Genome-Wide Association Study (GWAS) database focused on GM and CKD. For examination of potential causal connection from GM to CKD, a 2-way, 2-sample Mendelian randomization (MR) method was applied. Sensitivity analyses were utilized to scrutinize for heterogeneity, horizontal pleiotropy, MR outcomes resilience. Result from inverse variance weighting (IVW) method revealed that 10 microbiotas such as Porphyromonadaceae (OR = 1.351, 95% CI: 1.114-1.638, P = .002), Dorea (OR = 1.236, 95% CI: 1.040-1.468, P = .016), Ruminococcus torques group (OR = 1.290, 95% CI: 1.035-1.608, P = .024) are potential CKD risk factors. Five microbiotas, including the Prevotellaceae (OR = 0.814, 95% CI: 0.719-0.922, P = .001) are potential CKD protective factors. Sensitivity analyses reveal no horizontal pleiotropy or heterogeneity. Additionally, reverse MR results unveiled potential relation between CKD and disorders in 3 microbiotas, including Senegalimassilia. According to the investigation, MR method was employed to delve into reciprocal causal connection from GM to CKD. Our findings identified 15 types of GM causally linked to CKD, as well as CKD demonstrating causal associations with 3 types of GM. Further exploration of these associated GM types is hopeful to raise novel insights, for CKD preventing and early monitoring.
Collapse
Affiliation(s)
- Wen Wen Zhang
- First Clinical Medical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Jin Lin Huo
- College of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Pudong New Area, Shanghai, China
| | - Mei Di Xiao
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Ya Jie Xu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Jin Zhou
- First Clinical Medical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China
- Department of Traditional Chinese Medicine, Shenzhen Second People’s Hospital, Shenzhen Municipal Health Commission Traditional Chinese Medicine Key Specialty, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Wang X, Lang F, Liu D. High-Salt Diet and Intestinal Microbiota: Influence on Cardiovascular Disease and Inflammatory Bowel Disease. BIOLOGY 2024; 13:674. [PMID: 39336101 PMCID: PMC11429420 DOI: 10.3390/biology13090674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
Salt, or sodium chloride, is an essential component of the human diet. Recent studies have demonstrated that dietary patterns characterized by a high intake of salt can influence the abundance and diversity of the gut microbiota, and may play a pivotal role in the etiology and exacerbation of certain diseases, including inflammatory bowel disease and cardiovascular disease. The objective of this review is to synthesize the effects of elevated salt consumption on the gut microbiota, including its influence on gut microbial metabolites and the gut immune system. Additionally, this review will investigate the potential implications of these effects for the development of cardiovascular disease and inflammatory bowel disease. The findings of this study offer novel insights and avenues for the management of two common conditions with significant clinical implications.
Collapse
Affiliation(s)
- Xueyang Wang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.W.); (F.L.)
| | - Fuyuan Lang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.W.); (F.L.)
| | - Dan Liu
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.W.); (F.L.)
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
4
|
Akhlaghi E, Salari E, Mansouri M, Shafiei M, Kalantar-Neyestanaki D, Aghassi H, Fasihi Harandi M. Identification and comparison of intestinal microbial diversity in patients at different stages of hepatic cystic echinococcosis. Sci Rep 2024; 14:18912. [PMID: 39143364 PMCID: PMC11324937 DOI: 10.1038/s41598-024-70005-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024] Open
Abstract
There is a significant focus on the role of the host microbiome in different outcomes of human parasitic diseases, including cystic echinococcosis (CE). This study was conducted to identify the intestinal microbiome of patients with CE at different stages of hydatid cyst compared to healthy individuals. Stool samples from CE patients as well as healthy individuals were collected. The samples were divided into three groups representing various stages of hepatic hydatid cyst: active (CE1 and CE2), transitional (CE3), and inactive (CE4 and CE5). One family member from each group was selected to serve as a control. The gut microbiome of patients with different stages of hydatid cysts was investigated using metagenomic next-generation amplicon sequencing of the V3-V4 region of the 16S rRNA gene. In this study, we identified 4862 Operational Taxonomic Units from three stages of hydatid cysts in CE patients and healthy individuals with a combined frequency of 2,955,291. The most abundant genera observed in all the subjects were Blautia, Agathobacter, Faecalibacterium, Bacteroides, Bifidobacterium, and Prevotella. The highest microbial frequency was related to inactive forms of CE, and the lowest frequency was observed in the group with active forms. However, the lowest OTU diversity was found in patients with inactive cysts compared with those with active and transitional cyst stages. The genus Agatobacter had the highest OTU frequency. Pseudomonas, Gemella, and Ligilactobacillus showed significant differences among the patients with different stages of hydatid cysts. Additionally, Anaerostipes and Candidatus showed significantly different reads in CE patients compared to healthy individuals. Our findings indicate that several bacterial genera can play a role in the fate of hydatid cysts in patients at different stages of the disease.
Collapse
Affiliation(s)
- Elham Akhlaghi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Salari
- Department of Plant Protection, Faculty of Plant Production, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Mehdi Mansouri
- Department of Agricultural Biotechnology, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Shafiei
- Research Center for Hydatid Disease in Iran, Department of Surgery, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Kalantar-Neyestanaki
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Medical Microbiology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Aghassi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
5
|
Zhou M, Li X, Liu J, Wu Y, Tan Z, Deng N. Adenine's impact on mice's gut and kidney varies with the dosage administered and relates to intestinal microorganisms and enzyme activities. 3 Biotech 2024; 14:88. [PMID: 38406640 PMCID: PMC10884393 DOI: 10.1007/s13205-024-03959-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
This study aimed to investigate the effects of different dosages of adenine on intestinal microorganisms and enzyme activities, laying the experimental groundwork for subsequent exploration of the microbial mechanisms underlying diarrhea with kidney yang deficiency syndrome. Twenty-four mice were assigned to the following four groups: the control (NC) group, low-dosage adenine (NML) group, middle-dosage adenine (NMM) group, and high-dosage adenine (NMH) group. Mice in the NML, NMM, and NMH groups received 25 mg/(kg·d), 50 mg/(kg·d), and 100 mg/(kg·d) of adenine, respectively, 0.4 mL/each, once a day for 14 days. The NC group received 0.4 mL sterile water. Parameters including body weight, rectal temperature, intestinal microorganisms, enzyme activities, and microbial activity were measured. Results indicated that mice in the experimental group displayed signs of a poor mental state, curled up with their backs arched, and felt sleepy and lazy, with sparse fur that was easily shed, and damp bedding. Some mice showed fecal adhesion contamination in the perianal and tail areas. Dosage-dependent effects were observed, with decreased food intake, body weight, rectal temperature, and microbial activity and increased water intake and fecal water content. Enzyme activity analyses revealed significantly higher activities of protease, sucrase, amylase, and cellulase in intestinal contents and lactase, sucrase, amylase, and cellulase in the mucosa of the NMM group compared to those of other groups. Ultimately, the higher adenine dosage was associated with more pronounced symptoms of kidney yang deficiency syndrome, with 50 mg/kg adenine exhibiting the most substantial impact on the number of intestinal microbial colonies and enzyme activities.
Collapse
Affiliation(s)
- Mengsi Zhou
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| | - Xiaoya Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650000 China
| | - Jin Liu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| | - Yi Wu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| | - Zhoujin Tan
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| | - Na Deng
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| |
Collapse
|
6
|
Ryguła I, Pikiewicz W, Grabarek BO, Wójcik M, Kaminiów K. The Role of the Gut Microbiome and Microbial Dysbiosis in Common Skin Diseases. Int J Mol Sci 2024; 25:1984. [PMID: 38396663 PMCID: PMC10889245 DOI: 10.3390/ijms25041984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/28/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Dermatoses are an increasingly common problem, particularly in developed countries. The causes of this phenomenon include genetic factors and environmental elements. More and more scientific reports suggest that the gut microbiome, more specifically its dysbiosis, also plays an important role in the induction and progression of diseases, including dermatological diseases. The gut microbiome is recognised as the largest endocrine organ, and has a key function in maintaining human homeostasis. In this review, the authors will take a close look at the link between the gut-skin axis and the pathogenesis of dermatoses such as atopic dermatitis, psoriasis, alopecia areata, and acne. The authors will also focus on the role of probiotics in remodelling the microbiome and the alleviation of dermatoses.
Collapse
Affiliation(s)
- Izabella Ryguła
- Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Wojciech Pikiewicz
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Beniamin Oskar Grabarek
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Michał Wójcik
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| | - Konrad Kaminiów
- Collegium Medicum—Faculty of Medicine, WSB University, 41-300 Dabrowa Gornicza, Poland; (W.P.); (B.O.G.); (M.W.)
| |
Collapse
|
7
|
Zhao J, Zhao C, Xun T, Wang X, Wei S, Ye C, Zhang M, Guo D, Yang X. Huang Gan Formula Alleviates Systemic Inflammation and Uremia in Adenine-Induced Chronic Kidney Disease Rats May Associate with Modification of Gut Microbiota and Colonic Microenvironment. Drug Des Devel Ther 2024; 18:13-28. [PMID: 38205394 PMCID: PMC10777866 DOI: 10.2147/dddt.s421446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Purpose This study aims to investigate the effects of Huang Gan formula (HGF), a Chinese herbal prescription used for chronic kidney disease (CKD), on the regulation of the gut microbiota and colonic microenvironment of CKD. Methods CKD rats were induced by 150 mg/kg adenine gavage for 4 weeks, then orally treated with or without 3.6 g/kg or 7.2 g/kg of HGF for 8 weeks. The renal function and structure were analyzed by biochemical detection, hematoxylin and eosin, Masson's trichrome, Sirius red and immunochemical staining. Average fecal weight and number in the colon were recorded to assess colonic motility. Further, the changes in the gut microbiota and colonic microenvironment were evaluated by 16S rRNA sequencing, RT-PCR or immunofluorescence. The levels of inflammatory cytokines, uremic toxins, and NF-κB signaling pathway were detected by RT-PCR, ELISA, chloramine-T method or Western blotting. Redundancy analysis biplot and Spearman's rank correlation coefficient were used for correlation analysis. Results HGF significantly improved renal function and pathological injuries of CKD. HGF could improve gut microbial dysbiosis, protect colonic barrier and promote motility of colonic lumens. Further, HGF inhibited systemic inflammation through a reduction of TNF-α, IL-6, IL-1β, TGF-β1, and a suppression of NF-κB signaling pathway. The serum levels of the selected uremic toxins were also reduced by HGF treatment. Spearman correlation analysis suggested that high-dose HGF inhibited the overgrowth of bacteria that were positively correlated with inflammatory factors (eg, TNF-α) and uremic toxins (eg, indoxyl sulfate), whereas it promoted the proliferation of bacteria belonging to beneficial microbial groups and was positively correlated with the level of IL-10. Conclusion Our results suggest that HGF can improve adenine-induced CKD via suppressing systemic inflammation and uremia, which may associate with the regulations of the gut microbiota and colonic microenvironment.
Collapse
Affiliation(s)
- Jingqian Zhao
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Chenyu Zhao
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Tianrong Xun
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| | - Xiaokang Wang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| | - Sui Wei
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Chunxiao Ye
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| | - Mimi Zhang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| | - Dan Guo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| |
Collapse
|
8
|
Wang Y, Guo Z, Li J, Sui F, Dai W, Zhang W, Du H. Unraveling the differential perturbations of species-level functional profiling of gut microbiota among phases of methamphetamine-induced conditioned place preference. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110828. [PMID: 37459963 DOI: 10.1016/j.pnpbp.2023.110828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
The gut microbiome plays a significant role in methamphetamine addiction. Previous studies using short-read amplicon sequencing have described alterations in microbiota at the genus level and predicted function, in which taxonomic resolution is insufficient for accurate functional measurements. To address this limitation, we employed metagenome sequencing to intuitively associate species to functions of gut microbiota in methamphetamine-induced conditioned place preference. We observed differential perturbations of species-level functional profiling of the gut microbiota across phases of METH-induced CPP, with alterations in SCFA metabolism and bacterial motility at the acquisition phase and substance dependence-alcoholism pathway and amino acid metabolism at the extinction phase. Our findings suggest that reduced beneficial bacteria, i.e., Lactobacillus reuteri, contributed to the alteration of SCFA metabolism, while the increased abundance of Akkermansia muciniphila during the extinction phase may be associated with altered phenylalanine, tyrosine, and tryptophan metabolism and substance dependence pathway. Our study further supports the association between specific microbial taxa and METH-induced rewarding.
Collapse
Affiliation(s)
- Yinan Wang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China; School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhonghao Guo
- School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jing Li
- University of Science and Technology of China, Anhui, China
| | - Fang Sui
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenkui Dai
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenyong Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Hui Du
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
9
|
Altamura S, Pietropaoli D, Lombardi F, Del Pinto R, Ferri C. An Overview of Chronic Kidney Disease Pathophysiology: The Impact of Gut Dysbiosis and Oral Disease. Biomedicines 2023; 11:3033. [PMID: 38002033 PMCID: PMC10669155 DOI: 10.3390/biomedicines11113033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Chronic kidney disease (CKD) is a severe condition and a significant public health issue worldwide, carrying the burden of an increased risk of cardiovascular events and mortality. The traditional factors that promote the onset and progression of CKD are cardiometabolic risk factors like hypertension and diabetes, but non-traditional contributors are escalating. Moreover, gut dysbiosis, inflammation, and an impaired immune response are emerging as crucial mechanisms in the disease pathology. The gut microbiome and kidney disease exert a reciprocal influence commonly referred to as "the gut-kidney axis" through the induction of metabolic, immunological, and endocrine alterations. Periodontal diseases are strictly involved in the gut-kidney axis for their impact on the gut microbiota composition and for the metabolic and immunological alterations occurring in and reciprocally affecting both conditions. This review aims to provide an overview of the dynamic biological interconnections between oral health status, gut, and renal pathophysiology, spotlighting the dynamic oral-gut-kidney axis and raising whether periodontal diseases and gut microbiota can be disease modifiers in CKD. By doing so, we try to offer new insights into therapeutic strategies that may enhance the clinical trajectory of CKD patients, ultimately advancing our quest for improved patient outcomes and well-being.
Collapse
Affiliation(s)
- Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- PhD School in Medicine and Public Health, Center of Oral Diseases, Prevention and Translational Research—Dental Clinic, 67100 L’Aquila, Italy
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
| | - Davide Pietropaoli
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
- Center of Oral Diseases, Prevention and Translational Research—Dental Clinic, 67100 L’Aquila, Italy
| | - Francesca Lombardi
- Laboratory of Immunology and Immunopathology, Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Rita Del Pinto
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
- Unit of Internal Medicine and Nephrology, Center for Hypertension and Cardiovascular Prevention, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
- Unit of Internal Medicine and Nephrology, Center for Hypertension and Cardiovascular Prevention, San Salvatore Hospital, 67100 L’Aquila, Italy
| |
Collapse
|
10
|
Baima G, Ribaldone DG, Romano F, Aimetti M, Romandini M. The Gum-Gut Axis: Periodontitis and the Risk of Gastrointestinal Cancers. Cancers (Basel) 2023; 15:4594. [PMID: 37760563 PMCID: PMC10526746 DOI: 10.3390/cancers15184594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Periodontitis has been linked to an increased risk of various chronic non-communicable diseases, including gastrointestinal cancers. Indeed, dysbiosis of the oral microbiome and immune-inflammatory pathways related to periodontitis may impact the pathophysiology of the gastrointestinal tract and its accessory organs through the so-called "gum-gut axis". In addition to the hematogenous spread of periodontal pathogens and inflammatory cytokines, recent research suggests that oral pathobionts may translocate to the gastrointestinal tract through saliva, possibly impacting neoplastic processes in the gastrointestinal, liver, and pancreatic systems. The exact mechanisms by which oral pathogens contribute to the development of digestive tract cancers are not fully understood but may involve dysbiosis of the gut microbiome, chronic inflammation, and immune modulation/evasion, mainly through the interaction with T-helper and monocytic cells. Specifically, keystone periodontal pathogens, including Porphyromonas gingivalis and Fusobacterium nucleatum, are known to interact with the molecular hallmarks of gastrointestinal cancers, inducing genomic mutations, and promote a permissive immune microenvironment by impairing anti-tumor checkpoints. The evidence gathered here suggests a possible role of periodontitis and oral dysbiosis in the carcinogenesis of the enteral tract. The "gum-gut axis" may therefore represent a promising target for the development of strategies for the prevention and treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Giacomo Baima
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | | | - Federica Romano
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | - Mario Aimetti
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | - Mario Romandini
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0313 Oslo, Norway
| |
Collapse
|
11
|
Pantazi AC, Kassim MAK, Nori W, Tuta LA, Mihai CM, Chisnoiu T, Balasa AL, Mihai L, Lupu A, Frecus CE, Lupu VV, Chirila SI, Badescu AG, Hangan LT, Cambrea SC. Clinical Perspectives of Gut Microbiota in Patients with Chronic Kidney Disease and End-Stage Kidney Disease: Where Do We Stand? Biomedicines 2023; 11:2480. [PMID: 37760920 PMCID: PMC10525496 DOI: 10.3390/biomedicines11092480] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/26/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The gut microbiota (GM) plays a vital role in human health, with increasing evidence linking its imbalance to chronic kidney disease and end-stage kidney disease. Although the exact methods underlying kidney-GM crosstalk are not fully understood, interventions targeting GM were made and lay in three aspects: diagnostic, predictive, and therapeutic interventions. While these interventions show promising results in reducing uremic toxins and inflammation, challenges remain in the form of patient-specific GM variability, potential side effects, and safety concerns. Our understanding of GMs role in kidney disease is still evolving, necessitating further research to elucidate the causal relationship and mechanistic interactions. Personalized interventions focusing on specific GM signatures could enhance patient outcomes. However, comprehensive clinical trials are needed to validate these approaches' safety, efficacy, and feasibility.
Collapse
Affiliation(s)
| | | | - Wassan Nori
- College of Medicine, Mustansiriyah University, Baghdad 10052, Iraq;
| | - Liliana Ana Tuta
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Cristina Maria Mihai
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Tatiana Chisnoiu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Adriana Luminita Balasa
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Larisia Mihai
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Ancuta Lupu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Corina Elena Frecus
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
- Clinical Emergency Hospital of Constanta, 900591 Constanta, Romania
| | - Vasile Valeriu Lupu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Sergiu Ioachim Chirila
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
| | | | - Laurentiu-Tony Hangan
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
| | - Simona Claudia Cambrea
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania (L.A.T.)
| |
Collapse
|
12
|
Copur S, Peltek IB, Mutlu A, Tanriover C, Kanbay M. A new immune disease: systemic hypertension. Clin Kidney J 2023; 16:1403-1419. [PMID: 37664577 PMCID: PMC10469084 DOI: 10.1093/ckj/sfad059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Indexed: 09/05/2023] Open
Abstract
Systemic hypertension is the most common medical comorbidity affecting the adult population globally, with multiple associated outcomes including cerebrovascular diseases, cardiovascular diseases, vascular calcification, chronic kidney disease, metabolic syndrome and mortality. Despite advancements in the therapeutic field approximately one in every five adult patients with hypertension is classified as having treatment-resistant hypertension, indicating the need for studies to provide better understanding of the underlying pathophysiology and the need for more therapeutic targets. Recent pre-clinical studies have demonstrated the role of the innate and adaptive immune system including various cell types and cytokines in the pathophysiology of hypertension. Moreover, pre-clinical studies have indicated the potential beneficial effects of immunosuppressant medications in the control of hypertension. Nevertheless, it is unclear whether such pathophysiological mechanisms and therapeutic alternatives are applicable to human subjects, while this area of research is undoubtedly a rapidly growing field.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ibrahim B Peltek
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ali Mutlu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Cem Tanriover
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Department of Medicine, Section of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
13
|
Kim KR, Kim SM, Kim JH. A pilot study of alterations of the gut microbiome in canine chronic kidney disease. Front Vet Sci 2023; 10:1241215. [PMID: 37691637 PMCID: PMC10484476 DOI: 10.3389/fvets.2023.1241215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Gut dysbiosis has been noted in humans and animals with chronic kidney disease (CKD). However, little is known about the gut microbiome in canine patients with CKD. This study aimed to analyze and compare the gut microbiome profiles of healthy and CKD dogs, including differences in the gut microbiome between each CKD stage. Methods The study was conducted on 29 client-owned dogs who underwent physical examination, complete blood count (CBC), serum biochemistry, and urinalysis. The gut microbiome profile of healthy dogs (n = 10) and dogs with CKD (n = 19) was analyzed employing 16S rRNA sequencing. Results Significant differences were seen in the composition of the gut microbiome, with increased operational taxonomic units from the phylum Proteobacteria (p = 0.035), family Enterobacteriaceae (p < 0.001), and genus Enterococcus (p = 0.002) in dogs with CKD, and a decrease in the genus Ruminococcus (p = 0.007). Furthermore, an increase in both the progression of CKD and abundance of genus Klebsiella (Jonckheere-Terpstra test statistic value (JT) = 2.852, p = 0.004) and Clostridium (JT = 2.018, p = 0.044) was observed. Discussion Our study demonstrated that in dogs with CKD, the composition of the gut microbiome varied depending on the stage of CKD. Alterations in gut microbiome composition observed in CKD patients are characterized by an increase in proteolytic bacteria and a decrease in saccharolytic bacteria. These findings suggest specific gut microbiota could be targeted for clinical management of uremic dogs with CKD.
Collapse
Affiliation(s)
- Kyung-Ryung Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | | | - Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Sullivan VK, Appel LJ, Anderson CAM, Kim H, Unruh ML, Lash JP, Trego M, Sondheimer J, Dobre M, Pradhan N, Rao PS, Chen J, He J, Rebholz CM. Ultraprocessed Foods and Kidney Disease Progression, Mortality, and Cardiovascular Disease Risk in the CRIC Study. Am J Kidney Dis 2023; 82:202-212. [PMID: 37028638 PMCID: PMC10524102 DOI: 10.1053/j.ajkd.2023.01.452] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/22/2023] [Indexed: 04/09/2023]
Abstract
RATIONALE & OBJECTIVE Ultraprocessed foods are widely consumed in the United States and are associated with cardiovascular disease (CVD), mortality, and kidney function decline in the general population. We investigated associations between ultraprocessed food intake and chronic kidney disease (CKD) progression, all-cause mortality, and incident CVD in adults with chronic kidney disease (CKD). STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS Chronic Renal Insufficiency Cohort Study participants who completed baseline dietary questionnaires. EXPOSURE Ultraprocessed food intake (in servings per day) classified according to the NOVA system. OUTCOMES CKD progression (≥50% decrease in estimated glomerular filtration rate [eGFR] or initiation of kidney replacement therapy), all-cause mortality, and incident CVD (myocardial infarction, congestive heart failure, or stroke). ANALYTICAL APPROACH Cox proportional hazards models adjusted for demographic, lifestyle, and health covariates. RESULTS There were 1,047 CKD progression events observed during a median follow-up of 7 years. Greater ultraprocessed food intake was associated with higher risk of CKD progression (tertile 3 vs tertile 1, HR, 1.22; 95% CI, 1.04-1.42; P=0.01 for trend). The association differed by baseline kidney function, such that greater intake was associated with higher risk among people with CKD stages 1/2 (eGFR≥60mL/min/1.73m2; tertile 3 vs tertile 1, HR, 2.61; 95% CI, 1.32-5.18) but not stages 3a-5 (eGFR<60mL/min/1.73m2; P=0.003 for interaction). There were 1,104 deaths observed during a median follow-up of 14 years. Greater ultraprocessed food intake was associated with higher risk of mortality (tertile 3 vs tertile 1, HR, 1.21; 95% CI, 1.04-1.40; P=0.004 for trend). LIMITATIONS Self-reported diet. CONCLUSIONS Greater ultraprocessed food intake may be associated with CKD progression in earlier stages of CKD and is associated with higher risk of all-cause mortality in adults with CKD. PLAIN LANGUAGE SUMMARY Ultraprocessed foods are industrial formulations produced using ingredients and processes that are not commonly used in culinary preparations and contain few, if any, intact unprocessed foods. Ultraprocessed foods are widely consumed in the United States, and high intakes of such foods have been linked to cardiovascular disease, kidney disease, and mortality in the general population. In this study, we found that greater intake of ultraprocessed foods was associated with higher risk of kidney disease progression and mortality in adults with chronic kidney disease. Our findings suggest that patients with kidney disease may benefit from greater consumption of fresh, whole, and homemade or hand-prepared foods and fewer highly processed foods.
Collapse
Affiliation(s)
- Valerie K Sullivan
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence J Appel
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cheryl A M Anderson
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, California
| | - Hyunju Kim
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark L Unruh
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM
| | - James P Lash
- Department of Medicine, Division of Nephrology, University of Illinois at Chicago, Chicago, IL
| | - Marsha Trego
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - James Sondheimer
- Division of Nephrology and Hypertension, School of Medicine, Wayne State University, Detroit, MI
| | - Mirela Dobre
- Department of Medicine (Nephrology), Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Nishigandha Pradhan
- Department of Medicine (Nephrology), Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH
| | | | - Jing Chen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Casey M Rebholz
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
15
|
Kerem G, Yu X, Ismayi A, Teng B, Udduttula A, Liu C, Yu Z, Tohty D, Zhang JV, Ren PG. Small intestinal microbiota composition altered in obesity-T2DM mice with high salt fed. Sci Rep 2023; 13:8256. [PMID: 37217529 DOI: 10.1038/s41598-023-33909-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Obesity has become a global concern because of increasing the risk of many diseases. Alterations in human gut microbiota have been proven to be associated with obesity, yet the mechanism of how the microbiota are altered by high salt diet (HSD) remains obscure. In this study, the changes of Small Intestinal Microbiota (SIM) in obesity-T2DM mice were investigated. High-throughput sequencing was applied for the jejunum microbiota analysis. Results revealed that high salt intake (HS) could suppress the body weight (B.W.) in some extent. In addition, significant T2DM pathological features were revealed in high salt-high food diet (HS-HFD) group, despite of relatively lower food intake. High-throughput sequencing analysis indicated that the F/B ratio in HS intake groups increased significantly (P < 0.001), whereas beneficial bacteria, such as lactic acid or short chain fatty acid producing bacteria, were significantly decreased in HS-HFD group (P < 0.01 or P < 0.05). Furthermore, Halorubrum luteum were observed in small intestine for the first time. Above results preliminary suggested that in obesity-T2DM mice, high dietary salt could aggravate the imbalance of composition of SIM to unhealthy direction.
Collapse
Affiliation(s)
- Goher Kerem
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiangfang Yu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Aynur Ismayi
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bin Teng
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Anjaneyulu Udduttula
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chang Liu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Zhongjia Yu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Dilbar Tohty
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Jian V Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Key Laboratory of Nanomedicine, Shenzhen, China
| | - Pei-Gen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
16
|
Epigenetic Modifications Induced by the Gut Microbiota May Result from What We Eat: Should We Talk about Precision Diet in Health and Disease? Metabolites 2023; 13:metabo13030375. [PMID: 36984815 PMCID: PMC10051796 DOI: 10.3390/metabo13030375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Diet is currently considered one of the most important adjustable determinants of human health. The gut microbiota, the collection of microorganisms that inhabit (mainly) the distal bowel, has recently been shown to ensure critical physiological functions, such as immune, metabolic and neuropsychiatric. Many of these biological effects result from the production of bacterial metabolites that may target host cells, tissues and organs. In line with this rationale, epigenetics has brought new insights to our understanding of how environmental factors influence gene expression and, interestingly, gut microbiota metabolites have recently been proposed as novel and significant inducers of epigenetic modifications. Efforts have been dedicated to unveil how the production of specific metabolites influences the activity of epigenetic writers and erasers in order to establish a mechanistic link between gut microbiota, epigenetic modifications and health. Recent data is now evidencing how specific microbial metabolites shape the epigenetic landscape of eukaryotic cells, paving new avenues for innovative therapeutic strategies relying on diet-driven microbiota: epigenetic interactions. Herein is discussed the impact of diet on gut microbiota and the molecular mechanisms underlying microbiota–host interactions, highlighting the influence of diet on microbiota metabolome and how this may induce epigenetic modifications in host cells. Furthermore, it is hypothesized that epigenetics may be a key process transducing the effects of diet on gut microbiota with consequences for health and disease. Accordingly, innovating strategies of disease prevention based on a “precision diet”, a personalized dietary planning according to specific epigenetic targets, are discussed.
Collapse
|
17
|
Voroneanu L, Burlacu A, Brinza C, Covic A, Balan GG, Nistor I, Popa C, Hogas S, Covic A. Gut Microbiota in Chronic Kidney Disease: From Composition to Modulation towards Better Outcomes-A Systematic Review. J Clin Med 2023; 12:jcm12051948. [PMID: 36902734 PMCID: PMC10003930 DOI: 10.3390/jcm12051948] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND A bidirectional kidney-gut axis was described in patients with chronic kidney disease (CKD). On the one hand, gut dysbiosis could promote CKD progression, but on the other hand, studies reported specific gut microbiota alterations linked to CKD. Therefore, we aimed to systematically review the literature on gut microbiota composition in CKD patients, including those with advanced CKD stages and end-stage kidney disease (ESKD), possibilities to shift gut microbiota, and its impact on clinical outcomes. MATERIALS AND METHODS We performed a literature search in MEDLINE, Embase, Scopus, and Cochrane databases to find eligible studies using pre-specified keywords. Additionally, key inclusion and exclusion criteria were pre-defined to guide the eligibility assessment. RESULTS We retrieved 69 eligible studies which met all inclusion criteria and were analyzed in the present systematic review. Microbiota diversity was decreased in CKD patients as compared to healthy individuals. Ruminococcus and Roseburia had good power to discriminate between CKD patients and healthy controls (AUC = 0.771 and AUC = 0.803, respectively). Roseburia abundance was consistently decreased in CKD patients, especially in those with ESKD (p < 0.001). A model based on 25 microbiota dissimilarities had an excellent predictive power for diabetic nephropathy (AUC = 0.972). Several microbiota patterns were observed in deceased ESKD patients as compared to the survivor group (increased Lactobacillus, Yersinia, and decreased Bacteroides and Phascolarctobacterium levels). Additionally, gut dysbiosis was associated with peritonitis and enhanced inflammatory activity. In addition, some studies documented a beneficial effect on gut flora composition attributed to synbiotic and probiotic therapies. Large randomized clinical trials are required to investigate the impact of different microbiota modulation strategies on gut microflora composition and subsequent clinical outcomes. CONCLUSIONS Patients with CKD had an altered gut microbiome profile, even at early disease stages. Different abundance at genera and species levels could be used in clinical models to discriminate between healthy individuals and patients with CKD. ESKD patients with an increased mortality risk could be identified through gut microbiota analysis. Modulation therapy studies are warranted.
Collapse
Affiliation(s)
- Luminita Voroneanu
- Nephrology Department, Dialysis and Renal Transplant Center, “Dr. C.I. Parhon” University Hospital, 700503 Iasi, Romania
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
| | - Alexandru Burlacu
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
- Department of Interventional Cardiology, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Crischentian Brinza
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
- Department of Interventional Cardiology, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Andreea Covic
- Nephrology Department, Dialysis and Renal Transplant Center, “Dr. C.I. Parhon” University Hospital, 700503 Iasi, Romania
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
- Correspondence:
| | - Gheorghe G. Balan
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, St. 1 Spiridon Emergency County Hospital, 700111 Iasi, Romania
| | - Ionut Nistor
- Nephrology Department, Dialysis and Renal Transplant Center, “Dr. C.I. Parhon” University Hospital, 700503 Iasi, Romania
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
| | - Cristina Popa
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
| | - Simona Hogas
- Nephrology Department, Dialysis and Renal Transplant Center, “Dr. C.I. Parhon” University Hospital, 700503 Iasi, Romania
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
| | - Adrian Covic
- Nephrology Department, Dialysis and Renal Transplant Center, “Dr. C.I. Parhon” University Hospital, 700503 Iasi, Romania
- Faculty of Medicine, ‘Grigore T. Popa’ University of Medicine, 700115 Iasi, Romania
| |
Collapse
|
18
|
Hassan D, Zahra T, Kanaan G, Khan MU, Mushtaq K, Nashwan AJ, Hamid PF. The Impact of Gut Microbiome Constitution to Reduce Cardiovascular Disease Risk: A Systematic Review and Meta-Analysis. Curr Probl Cardiol 2023; 48:101459. [PMID: 36261101 DOI: 10.1016/j.cpcardiol.2022.101459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 01/04/2023]
Affiliation(s)
- Danial Hassan
- Department of Healthcare Profession ((DHP), Ministry of Public Health, Qatar / ECPE, Harvard TH Chan School of Public Health, USA; ECPE, Harvard TH Chan School of Public Health, Boston, MA.
| | - Tatheer Zahra
- Department of Pediatrics, Allied Hospital, Faisalabad, Pakistan
| | - Ghid Kanaan
- California Institute of Behavioral Neurosciences and Psychology, USA
| | | | - Kamran Mushtaq
- Department of Gastroenterology Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | | | | |
Collapse
|
19
|
Ardalan M, Ahmadian E, Hosseiniyan Khatibi SM, Rahbar Saadat Y, Bastami M, Bagheri Y, Zununi Vahed F, Shoja MM, Zununi Vahed S. Microbiota and glomerulonephritis: An immunological point of view. Am J Med Sci 2022; 364:695-705. [PMID: 35870511 DOI: 10.1016/j.amjms.2022.05.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 03/26/2022] [Accepted: 05/05/2022] [Indexed: 01/25/2023]
Abstract
Glomerular injury is the major cause of chronic kidney diseases (CKD) worldwide and is characterized by proteinuria. Glomerulonephritis (GN) has a wide spectrum of etiologies, the intensity of glomerular damage, histopathology, and clinical outcomes that can be associated with the landscape of the nephritogenic immune response. Beyond impaired immune responses and genetic factors, recent evidence indicates that microbiota can be contributed to the pathogenesis of GN and patients' outcomes by impacting many aspects of the innate and adaptive immune systems. It is still unknown whether dysbiosis induces GN or it is a secondary effect of the disease. Several factors such as drugs and nutritional problems can lead to dysbiosis in GN patients. It has been postulated that gut dysbiosis activates immune responses, promotes a state of systemic inflammation, and produces uremic toxins contributing to kidney tissue inflammation, apoptosis, and subsequent proteinuric nephropathy. In this review, the impact of gastrointestinal tract (GI) microbiota on the pathogenesis of the primary GN will be highlighted. The application of therapeutic interventions based on the manipulation of gut microbiota with special diets and probiotic supplementation can be effective in GN.
Collapse
Affiliation(s)
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Milad Bastami
- Non-communicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Yasin Bagheri
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammadali M Shoja
- Clinical Academy of Teaching and Learning, Ross University School of Medicine, Miramar, FL, USA
| | | |
Collapse
|
20
|
Holle J, Bartolomaeus H, Löber U, Behrens F, Bartolomaeus TU, Anandakumar H, Wimmer MI, Vu DL, Kuhring M, Brüning U, Maifeld A, Geisberger S, Kempa S, Schumacher F, Kleuser B, Bufler P, Querfeld U, Kitschke S, Engler D, Kuhrt LD, Drechsel O, Eckardt KU, Forslund SK, Thürmer A, McParland V, Kirwan JA, Wilck N, Müller D. Inflammation in Children with CKD Linked to Gut Dysbiosis and Metabolite Imbalance. J Am Soc Nephrol 2022; 33:2259-2275. [PMID: 35985814 PMCID: PMC9731629 DOI: 10.1681/asn.2022030378] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/29/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND CKD is characterized by a sustained proinflammatory response of the immune system, promoting hypertension and cardiovascular disease. The underlying mechanisms are incompletely understood but may be linked to gut dysbiosis. Dysbiosis has been described in adults with CKD; however, comorbidities limit CKD-specific conclusions. METHODS We analyzed the fecal microbiome, metabolites, and immune phenotypes in 48 children (with normal kidney function, CKD stage G3-G4, G5 treated by hemodialysis [HD], or kidney transplantation) with a mean±SD age of 10.6±3.8 years. RESULTS Serum TNF-α and sCD14 were stage-dependently elevated, indicating inflammation, gut barrier dysfunction, and endotoxemia. We observed compositional and functional alterations of the microbiome, including diminished production of short-chain fatty acids. Plasma metabolite analysis revealed a stage-dependent increase of tryptophan metabolites of bacterial origin. Serum from patients on HD activated the aryl hydrocarbon receptor and stimulated TNF-α production in monocytes, corresponding to a proinflammatory shift from classic to nonclassic and intermediate monocytes. Unsupervised analysis of T cells revealed a loss of mucosa-associated invariant T (MAIT) cells and regulatory T cell subtypes in patients on HD. CONCLUSIONS Gut barrier dysfunction and microbial metabolite imbalance apparently mediate the proinflammatory immune phenotype, thereby driving the susceptibility to cardiovascular disease. The data highlight the importance of the microbiota-immune axis in CKD, irrespective of confounding comorbidities.
Collapse
Affiliation(s)
- Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Behrens
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Institute of Physiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Theda U.P. Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Harithaa Anandakumar
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz I. Wimmer
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Tübingen, Tübingen, Germany
| | - Dai Long Vu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Metabolomics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Kuhring
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrike Brüning
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Metabolomics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Andras Maifeld
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sabrina Geisberger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- The Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- The Berlin Institute for Medical Systems Biology, Berlin, Germany
| | | | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Querfeld
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Stefanie Kitschke
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Denise Engler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Leonard D. Kuhrt
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sofia K. Forslund
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Andrea Thürmer
- MF2 Genome Sequencing, Robert Koch Institute, Berlin, Germany
| | - Victoria McParland
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jennifer A. Kirwan
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Metabolomics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Dominik Müller
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
21
|
Are South African Wild Foods the Answer to Rising Rates of Cardiovascular Disease? DIVERSITY 2022. [DOI: 10.3390/d14121014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The rising burden of cardiovascular disease in South Africa gives impetus to managerial changes, particularly to the available foods in the market. Since there are many economically disadvantaged groups in urban societies who are at the forefront of the CVD burden, initiatives to make healthier foods available should focus on affordability in conjunction with improved phytochemical diversity to incentivize change. The modern obesogenic diet is deficient in phytochemicals that are protective against the metabolic products of sugar metabolism, i.e., inflammation, reactive oxygen species and mitochondrial fatigue, whereas traditional southern African food species have high phytochemical diversity and are also higher in soluble dietary fibres that modulate the release of sugars from starches, nurture the microbiome and produce digestive artefacts that are prophylactic against cardiovascular disease. The examples of indigenous southern African food species with high horticultural potential that can be harvested sustainably to feed a large market of consumers include: Aloe marlothii, Acanthosicyos horridus, Adansonia digitata, Aloe ferox, Amaranthus hybridus, Annesorhiza nuda, Aponogeton distachyos, Bulbine frutescens, Carpobrotus edulis, Citrullus lanatus, Dioscorea bulbifera, Dovyalis caffra, Eleusine coracana, Lagenaria siceraria, Mentha longifolia, Momordica balsamina, Pelargonium crispum, Pelargonium sidoides, Pennisetum glaucum, Plectranthus esculentus, Schinziophyton rautanenii, Sclerocarya birrea, Solenostemon rotundifolius, Talinum caffrum, Tylosema esculentum, Vigna unguiculata and Vigna subterranea. The current review explains the importance of phytochemical diversity in the human diet, it gives a lucid explanation of phytochemical groups and links the phytochemical profiles of these indigenous southern African foods to their protective effects against cardiovascular disease.
Collapse
|
22
|
Zhou W, Wu WH, Si ZL, Liu HL, Wang H, Jiang H, Liu YF, Alolga RN, Chen C, Liu SJ, Bian XY, Shan JJ, Li J, Tan NH, Zhang ZH. The gut microbe Bacteroides fragilis ameliorates renal fibrosis in mice. Nat Commun 2022; 13:6081. [PMID: 36241632 PMCID: PMC9568537 DOI: 10.1038/s41467-022-33824-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/03/2022] [Indexed: 12/24/2022] Open
Abstract
Renal fibrosis is an inevitable outcome of various manifestations of progressive chronic kidney diseases (CKD). The need for efficacious treatment regimen against renal fibrosis can therefore not be overemphasized. Here we show a novel protective role of Bacteroides fragilis (B. fragilis) in renal fibrosis in mice. We demonstrate decreased abundance of B. fragilis in the feces of CKD patients and unilateral ureteral obstruction (UUO) mice. Oral administration of live B. fragilis attenuates renal fibrosis in UUO and adenine mice models. Increased lipopolysaccharide (LPS) levels are decreased after B. fragilis administration. Results of metabolomics and proteomics studies show decreased level of 1,5-anhydroglucitol (1,5-AG), a substrate of SGLT2, which increases after B. fragilis administration via enhancement of renal SGLT2 expression. 1,5-AG is an agonist of TGR5 that attenuates renal fibrosis by inhibiting oxidative stress and inflammation. Madecassoside, a natural product found via in vitro screening promotes B. fragilis growth and remarkably ameliorates renal fibrosis. Our findings reveal the ameliorative role of B. fragilis in renal fibrosis via decreasing LPS and increasing 1,5-AG levels.
Collapse
Affiliation(s)
- Wei Zhou
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wen-hui Wu
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zi-lin Si
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hui-ling Liu
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanyu Wang
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong Jiang
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ya-fang Liu
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Raphael N. Alolga
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng Chen
- grid.412632.00000 0004 1758 2270Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi-jia Liu
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xue-yan Bian
- grid.13402.340000 0004 1759 700XNingbo Hospital of Zhejiang University, Ningbo, China
| | - Jin-jun Shan
- grid.410745.30000 0004 1765 1045Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Li
- grid.254147.10000 0000 9776 7793School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ning-hua Tan
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhi-hao Zhang
- grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
23
|
Lacticaseibacillus rhamnosus Fmb14 prevents purine induced hyperuricemia and alleviate renal fibrosis through gut-kidney axis. Pharmacol Res 2022; 182:106350. [PMID: 35843568 DOI: 10.1016/j.phrs.2022.106350] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 11/22/2022]
Abstract
Hyperuricemia is a critical threat to human health, and conventional medical treatment only aims to treat acute gouty arthritis. Purine diet-mediated chronic hyperuricemia and related syndromes are neglected in clinical therapeutics. In this study, the prevention ability of Lacticaseibacillus rhamnosus Fmb14, screened from Chinese yogurt, was evaluated in chronic purine-induced hyperuricemia (CPH) mice. After 12 weeks of Fmb14 administration, serum uric acid (SUA) in CPH mice decreased by 36.8 %, from 179.1 to 113.2 µmol/L, and the mortality rate decreased from 30 % to 10 %. The prevention role of Fmb14 in CPH was further investigated, and the reduction of uric acid by Fmb14 was attributed to the reduction of XOD (xanthine oxidase) in the liver and URAT1 in the kidney, as well the promotion of ABCG2 in the colon. Fmb14 administration Increased ZO-1 and Occludin expression in the colon and decreased fibrosis degree in the kidney indicated that Fmb14 administration had preventive effects through the gut-kidney axis in CPH. In specific, Fmb14 administration upregulated the diversity of gut microbiota, increased short-chain fatty acids (SCFA) by 35 % in colon materials and alleviated the inflammatory response by reducing biomarkers levels of IL-1β, IL-18 and TNF-α at 11.6 %, 21.7 % and 26.5 % in serum, compared to CPH group, respectively. Additionally, 16 S rRNA sequencing showed 31.5 % upregulation of Prevotella, 20.5 % and 21.6 % downregulation of Ruminococcus and Suterella at the genus level, which may be a new gut microbial marker in hyperuricemia. In conclusion, Fmb14 ameliorated CPH through the gut-kidney axis, suggesting a new strategy to prevent hyperuricemia.
Collapse
|
24
|
Kanbay M, Copur S, Demiray A, Sag AA, Covic A, Ortiz A, Tuttle KR. Fatty kidney: A possible future for chronic kidney disease research. Eur J Clin Invest 2022; 52:e13748. [PMID: 35040119 DOI: 10.1111/eci.13748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Metabolic syndrome is a growing twenty-first century pandemic associated with multiple clinical comorbidities ranging from cardiovascular diseases, non-alcoholic fatty liver disease and polycystic ovary syndrome to kidney dysfunction. A novel area of research investigates the concept of fatty kidney in the pathogenesis of chronic kidney disease, especially in patients with diabetes mellitus or metabolic syndrome. AIM To review the most updated literature on fatty kidney and provide future research, diagnostic and therapeutic perspectives on a disease increasingly affecting the contemporary world. MATERIALS AND METHOD We performed an extensive literature search through three databases including Embase (Elsevier) and the Cochrane Central Register of Controlled Trials (Wiley) and PubMed/Medline Web of Science in November 2021 by using the following terms and their combinations: 'fatty kidney', 'ectopic fat', 'chronic kidney disease', 'cardiovascular event', 'cardio-metabolic risk', 'albuminuria' and 'metabolic syndrome'. Each study has been individually assessed by the authors. RESULTS Oxidative stress and inflammation, Klotho deficiency, endoplasmic reticulum stress, mitochondrial dysfunction and disruption of cellular energy balance appear to be the main pathophysiological mechanisms leading to tissue damage following fat accumulation. Despite the lack of large-scale comprehensive studies in this novel field of research, current clinical trials demonstrate fatty kidney as an independent risk factor for the development of chronic kidney disease and cardiovascular events. CONCLUSION The requirement for future studies investigating the pathophysiology, clinical outcomes and therapeutics of fatty kidney is clear.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alan A Sag
- Division of Vascular and Interventional Radiology, Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Adrian Covic
- Department of Nephrology, Grigore T. Popa' University of Medicine, Iasi, Romania
| | - Alberto Ortiz
- Department of Medicine, Universidad Autonoma de Madrid and IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Kathherine R Tuttle
- Division of Nephrology, University of Washington, Seattle, Washington, USA.,Providence Medical Research Center, Providence Health Care, Spokane, Washington, USA
| |
Collapse
|
25
|
Intestinal Carriage of Extended-spectrum β-lactamase and Carbapenemase-producing Enterobacteriaceae in Hemodialysis Patients. Jundishapur J Microbiol 2022. [DOI: 10.5812/jjm-118946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Gastrointestinal colonization with resistant pathogens is significant because they could be easily transmitted to other hosts or spread to different microbiota through mobile genetic elements. Objectives: This study assessed the prevalence of fecal carriage of extended-spectrum β-lactamase-producing and carbapenemase-producing Enterobacteriaceae (ESBL-E and CPE, respectively) among hemodialysis patients and the factors affecting its occurrence in a hospital in Tehran. Methods: From January 2018 to May 2019, 150 hemodialysis patients referred to a hospital in Tehran were sampled in this study. Stool samples of the patients were diluted in saline and cultured on MacConkey agar plates containing cefotaxime, ceftazidime, imipenem, and meropenem discs. The clinical data were analyzed to identify the risk factors using a logistic regression model. Results: The colonization rate of ESBL-E was 48.6%, while only 2% of patients were identified as the carriers of CPE (3 of 150). A higher prevalence rate was obtained for intestinal carriage of ESBL-E among hemodialysis patients aged 18 to 42 years using multivariate analysis. The prevalence rate of multidrug-resistant isolates was 73.8%. The blaCTX-M1 gene was identified as the most prevalent ESBL gene. Among carbapenemase-encoding genes, blaKPC and blaoxa-48 were found in 12 and two isolates, respectively. Conclusions: These results demonstrated a high prevalence rate of ESBLs among hemodialysis patients, although this rate was low for carbapenemases. Therefore, more control measures should be taken in hospitals to prevent the spread of antibiotic resistance genes in healthcare settings.
Collapse
|
26
|
Li M, Wei L, Sun J, Zhu Q, Yang H, Zhang Y, Zhang C, Xi L, Zhao R, Du X. Association of gut microbiota with idiopathic membranous nephropathy. BMC Nephrol 2022; 23:164. [PMID: 35488230 PMCID: PMC9055735 DOI: 10.1186/s12882-022-02797-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background The prevalence of idiopathic membranous nephropathy (IMN) is increasing worldwide and the gut microbiota is recognized to play a role in its pathology. The aim of this study was to understand the involvement of the gut–kidney axis in IMN by analyzing the composition of the gut microbiota of biopsy-proven IMN patients compared with healthy controls (HC). Methods Fecal samples from 30 patients with IMN diagnosed by renal biopsy and 30 healthy co-residents (control group) were collected for analysis in the Nephrology Department of the Second Affiliated Hospital of Harbin Medical University. The microbiota composition was analyzed by a 16S rRNA microbial profiling approach. Results The results indicated that the α- and β-diversity of IMN patients differed significantly from those of the HC groups (P < 0.05). At the phylum level, IMN patients showed an increased abundance of Proteobacteria but a reduced abundance of Bacteroidota compared with the HC group. Actinobacteriota abundance showed a strong negative correlation with the estimated glomerular filtration rate. At the genus level, Faecalibacterium, Agathobacter, and Bacteroides were less abundant in the IMN group than in the HC group (LDA score > 2). Abundant bacterial functions related to lipid metabolism were observed among IMN group. Conclusion Patients with IMN appear to have an altered gut microbiome, which could provide reference for future research on the interaction mechanism between the intestinal flora and IMN. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02797-5.
Collapse
Affiliation(s)
- Mengfei Li
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Lixue Wei
- Department of Nephrology, Jiaozuo People's Hospital, 267 Jiefang Middle Road, Henan, Jiaozuo, 454000, China
| | - Jing Sun
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Qianshen Zhu
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - He Yang
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Yu Zhang
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Chao Zhang
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Lin Xi
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Rong Zhao
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China
| | - Xuanyi Du
- Department of Nephrology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Ave, Harbin, Heilongjiang, 150086, China.
| |
Collapse
|
27
|
Ertuglu LA, Demiray A, Afsar B, Ortiz A, Kanbay M. The Use of Healthy Eating Index 2015 and Healthy Beverage Index for Predicting and Modifying Cardiovascular and Renal Outcomes. Curr Nutr Rep 2022; 11:526-535. [PMID: 35476188 DOI: 10.1007/s13668-022-00415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW With the wide recognition of the importance of dietary patterns rather than isolated nutrient groups on health outcomes, numerous diet quality indices have been designed to evaluate the overall food intake quality in the last two decades. RECENT FINDINGS The newest version of the Healthy Eating Index (HEI), HEI-2015, is a diet quality index that measures adherence to the recommendations of the 2015-2020 Dietary Guidelines for Americans. While the key nutrient groups are included in most diet quality indices, differences in other components and the scoring system differentiate HEI. The Healthy Beverage Index (HBI) was recently introduced. Previous literature has confirmed the association of the older versions of HEI with metabolic syndrome, inflammatory markers, and negative health outcomes including cardiovascular disease, type 2 diabetes mellitus, chronic kidney disease, and all-cause mortality. This review presents the existing evidence on the association of HEI-2015 and HBI with health markers and long-term outcome, provides guidance on their use, and identifies persisting challenges such as the development of simple, unified, and objective tools to characterize healthy diets in routine clinical practice.
Collapse
Affiliation(s)
- Lale A Ertuglu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Baris Afsar
- Division of Nephrology, Department of Internal Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Alberto Ortiz
- Department of Medicine, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, 34010, Istanbul, Turkey.
| |
Collapse
|
28
|
Vijay A, Valdes AM. Role of the gut microbiome in chronic diseases: a narrative review. Eur J Clin Nutr 2022; 76:489-501. [PMID: 34584224 PMCID: PMC8477631 DOI: 10.1038/s41430-021-00991-6] [Citation(s) in RCA: 206] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Amrita Vijay
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, The University of Nottingham, Nottingham, UK.
| | - Ana M Valdes
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, The University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| |
Collapse
|
29
|
Huang Y, Xin W, Xiong J, Yao M, Zhang B, Zhao J. The Intestinal Microbiota and Metabolites in the Gut-Kidney-Heart Axis of Chronic Kidney Disease. Front Pharmacol 2022; 13:837500. [PMID: 35370631 PMCID: PMC8971625 DOI: 10.3389/fphar.2022.837500] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Emerging evidences demonstrate the involvement of gut microbiota in the progression of chronic kidney disease (CKD) and CKD-associated complications including cardiovascular disease (CVD) and intestinal dysfunction. In this review, we discuss the interactions between the gut, kidney and heart in CKD state, and elucidate the significant role of intestinal microbiota in the gut-kidney-heart axis hypothesis for the pathophysiological mechanisms of these diseases, during which process mitochondria may serve as a potential therapeutic target. Dysregulation of this axis will lead to a vicious circle, contributing to CKD progression. Recent studies suggest novel therapies targeting gut microbiota in the gut-kidney-heart axis, including dietary intervention, probiotics, prebiotics, genetically engineered bacteria, fecal microbiota transplantation, bacterial metabolites modulation, antibiotics, conventional drugs and traditional Chinese medicine. Further, the identification of specific microbial communities and their corresponding pathophysiological metabolites and the illumination of the gut-kidney-heart axis may contribute to innovative basic research, clinical trials and therapeutic strategies against CKD progression and uremic complications in CKD patients.
Collapse
|
30
|
The Bidirectional Signal Communication of Microbiota-Gut-Brain Axis in Hypertension. Int J Hypertens 2022; 2021:8174789. [PMID: 34970454 PMCID: PMC8714396 DOI: 10.1155/2021/8174789] [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: 08/09/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
Hypertension is a critical risk factor of cardiovascular diseases. A new concept of microbiota-gut-brain axis has been established recently, mediating the bidirectional communication between the gut and its microbiome and the brain. Alterations in bidirectional interactions are believed to be involved in the blood pressure regulation. Neuroinflammation and increased sympathetic outflow act as the descending innervation signals from the brain. Increased sympathetic activation plays a recognized role in the genesis of hypertension. The present evidence demonstrates that gut dysbiosis is associated with central nervous system neuroinflammation. However, how the gut influences the brain remains unclear. We reviewed the roles of neuroinflammation and gut microbiota and their interactions in the pathogenesis of hypertension and described the ascending signaling mechanisms behind the microbiota-gut-brain axis in detail. Additionally, the innovative prohypertensive mechanisms of dietary salt through the microbiota-gut-brain axis are summarized. The bidirectional communication mechanisms were proposed for the first time that the descending signals from the brain and the ascending connections from the gut form a vicious circle of hypertension progression, acting as a premise for hypertension therapy.
Collapse
|
31
|
Role of the microbiota in hypertension and antihypertensive drug metabolism. Hypertens Res 2021; 45:246-253. [PMID: 34887530 DOI: 10.1038/s41440-021-00804-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/17/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that the gut microbiota plays an important role in the development and pathogenesis of hypertension. Dysbiosis, an imbalance in the composition and function of the gut microbiota, was shown to be associated with hypertension in both animal models and humans. In this review, we provide insights into host-microbiota interactions and summarize the evidence supporting the importance of the microbiota in blood pressure (BP) regulation. Metabolites produced by the gut microbiota, especially short-chain fatty acids (SCFAs), modulate BP and vascular responses. Harmful gut-derived metabolites, such as trimethylamine N-oxide and several uremic toxins, exert proatherosclerotic, prothrombotic, and proinflammatory effects. High-salt intake alters the composition of the microbiota, and this microbial alteration contributes to the pathogenesis of salt-sensitive hypertension. In addition, the microbiota may impact the metabolism of drugs and steroid hormones in the host. The drug-metabolizing activities of the microbiota affect the pharmacokinetic parameters of antihypertensive drugs and contribute to the pathogenesis of licorice-induced pseudohyperaldosteronism. Furthermore, the oral microbiota plays a role in BP regulation by producing nitric oxide, which lowers BP via its vasodilatory effects. Thus, antihypertensive intervention strategies targeting the microbiota, such as the use of prebiotics, probiotics, and postbiotics (e.g., SCFAs), are considered new therapeutic options for the treatment of hypertension.
Collapse
|
32
|
Sadgrove NJ, Padilla-González GF, Leuner O, Melnikovova I, Fernandez-Cusimamani E. Pharmacology of Natural Volatiles and Essential Oils in Food, Therapy, and Disease Prophylaxis. Front Pharmacol 2021; 12:740302. [PMID: 34744723 PMCID: PMC8566702 DOI: 10.3389/fphar.2021.740302] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022] Open
Abstract
This commentary critically examines the modern paradigm of natural volatiles in 'medical aromatherapy', first by explaining the semantics of natural volatiles in health, then by addressing chemophenetic challenges to authenticity or reproducibility, and finally by elaborating on pharmacokinetic and pharmacodynamic processes in food, therapy, and disease prophylaxis. Research over the last 50 years has generated substantial knowledge of the chemical diversity of volatiles, and their strengths and weaknesses as antimicrobial agents. However, due to modest in vitro outcomes, the emphasis has shifted toward the ability to synergise or potentiate non-volatile natural or pharmaceutical drugs, and to modulate gene expression by binding to the lipophilic domain of mammalian cell receptors. Because essential oils and natural volatiles are small and lipophilic, they demonstrate high skin penetrating abilities when suitably encapsulated, or if derived from a dietary item they bioaccumulate in fatty tissues in the body. In the skin or body, they may synergise or drive de novo therapeutic outcomes that range from anti-inflammatory effects through to insulin sensitisation, dermal rejuvenation, keratinocyte migration, upregulation of hair follicle bulb stem cells or complementation of anti-cancer therapies. Taking all this into consideration, volatile organic compounds should be examined as candidates for prophylaxis of cardiovascular disease. Considering the modern understanding of biology, the science of natural volatiles may need to be revisited in the context of health and nutrition.
Collapse
Affiliation(s)
| | | | - Olga Leuner
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Ingrid Melnikovova
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Eloy Fernandez-Cusimamani
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Prague, Czech Republic
| |
Collapse
|
33
|
Wu Y, Xu H, Tu X, Gao Z. The Role of Short-Chain Fatty Acids of Gut Microbiota Origin in Hypertension. Front Microbiol 2021; 12:730809. [PMID: 34650536 PMCID: PMC8506212 DOI: 10.3389/fmicb.2021.730809] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertension is a significant risk factor for cardiovascular and cerebrovascular diseases, and its development involves multiple mechanisms. Gut microbiota has been reported to be closely linked to hypertension. Short-chain fatty acids (SCFAs)-the metabolites of gut microbiota-participate in hypertension development through various pathways, including specific receptors, immune system, autonomic nervous system, metabolic regulation and gene transcription. This article reviews the possible mechanisms of SCFAs in regulating blood pressure and the prospects of SCFAs as a target to prevent and treat hypertension.
Collapse
Affiliation(s)
- Yeshun Wu
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Hongqing Xu
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xiaoming Tu
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Zhenyan Gao
- Department of Cardiology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| |
Collapse
|
34
|
Yang J, Chen W, Sun Y, Liu J, Zhang W. Effects of cadmium on organ function, gut microbiota and its metabolomics profile in adolescent rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 222:112501. [PMID: 34265528 DOI: 10.1016/j.ecoenv.2021.112501] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 06/13/2023]
Abstract
Cadmium (Cd) exposure in adult animals can result in multi-organ damages and gut microbiota disturbance. However, Cd's consequences on health and gut microbiota during adolescence are obscure. In the present study, three-week-old SD rats were exposed to Cd at doses of 0, 0.25, 1, and 4 mg/kg body weight for eight weeks, and the changes of liver, kidney, and ovary function, as well as gut microbiota and its metabolomics profile, were analyzed. After transplantation of fecal bacteria from the 4 mg/kg Cd-treated group into age-matched rats (4 mg/kg-Cd recipients), the organ function and inflammatory reaction were evaluated. The results indicated that Cd perturbed gut microbiota composition, significantly decreased the abundance of Prevotella and Lachnoclostridium but increased Escherichia coli_Shigella. The fecal metabolome profile was altered and was closely correlated with some specific genera. These changes were accompanied by the inflammatory response, dyslipidemia, kidney dysfunction, and abnormal estrogen level. In 4 mg/kg-Cd recipients, the serum triglyceride (TG), lipopolysaccharide (LPS), and inflammatory cytokines were increased with the expressions of IL-1β, IL-6, TNF-α genes up-regulated in liver and kidney. Overall, this study demonstrated that Cd exposure during adolescence could cause disturbance of gut microbiota, dysfunction of liver, kidney, and ovary, which may be correlated with the activation of Cd-induced inflammatory response.
Collapse
Affiliation(s)
- Jinsong Yang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyan Road No. 1, Minhou Coudslanty, Fuzhou 350108, China
| | - Wei Chen
- Department for Prevention and Control of Infectious Diseases, Fujian Center for Disease Control and Prevention, Jintai Road No. 76, Fuzhou 350001, China
| | - Yi Sun
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyan Road No. 1, Minhou Coudslanty, Fuzhou 350108, China
| | - Jin Liu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyan Road No. 1, Minhou Coudslanty, Fuzhou 350108, China
| | - Wenchang Zhang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Xueyan Road No. 1, Minhou Coudslanty, Fuzhou 350108, China.
| |
Collapse
|
35
|
Liu L, Su J, Li R, Luo F. Changes in Intestinal Flora Structure and Metabolites Are Associated With Myocardial Fibrosis in Patients With Persistent Atrial Fibrillation. Front Nutr 2021; 8:702085. [PMID: 34497820 PMCID: PMC8419273 DOI: 10.3389/fnut.2021.702085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background: The occurrence of atrial fibrillation is often accompanied by myocardial fibrosis. An increasing number of studies have shown that intestinal flora is involved in the occurrence and development of a variety of cardiovascular diseases. This study explores the relationship between changes in the structure and function of intestinal flora and the progression of myocardial fibrosis in patients with persistent atrial fibrillation. Methods: Serum and stool samples were collected from 10 healthy people and 10 patients with persistent atrial fibrillation (PeAF), and statistical analyses were performed on the subjects' clinical baseline conditions. ELISA was used to measure the levels of carboxy-terminal telopeptide of type I collagen (CTX-I), propeptide of type I procollagen (PICP), procollagen III N-terminal propeptide (PIIINP), fibroblast growth factor-23 (FGF-23), and transforming growth factor-beta 1 (TGF-β1) in serum. Through 16S rRNA sequencing technology, the structural composition of the intestinal flora was detected and analyzed. In addition, metabolomics data were analyzed to determine the differences in the metabolites produced by the intestinal flora of the subjects. Results: By comparing the baseline data of the subjects, it was found that compared with those of the control group, the levels of creatinine (CRE) and serum uric acid (SUA) in the serum of PeAF patients were significantly increased. In addition, we found that the levels of CTX-I, PICP, PIIINP, and TGF-β1 in the serum of PeAF patients were significantly higher than those of the control group subjects. Although the control and PeAF groups exhibited no significant differences in the α diversity index, there were significant differences in the β diversity indexes (Bray-Curtis, weighted UniFrac and Anosim). At the phylum, family and species levels, the community structure and composition of the intestinal flora of the control group and those of the PeAF group showed significant differences. In addition, the compositions of the intestinal metabolites in the two different groups of people were significantly different. They were correlated considerably with PIIINP and specific communities in the intestinal flora. Conclusion: Pathologically, PeAF patients may have a higher risk of myocardial fibrosis. Systematically, abnormal changes in the structure and composition of the intestinal flora in PeAF patients may lead to differences in intestinal metabolites, which are involved in the process of myocardial fibrosis through metabolite pathways.
Collapse
Affiliation(s)
- Langsha Liu
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Su
- Department of Medical Administration, Zhuzhou Central Hospital, Zhuzhou, China
| | - Rui Li
- Operating Theatre, Zhuzhou Central Hospital, Zhuzhou, China
| | - Fanyan Luo
- Department of Cardiac Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
36
|
Rodrigues FG, Ormanji MS, Heilberg IP, Bakker SJL, de Borst MH. Interplay between gut microbiota, bone health and vascular calcification in chronic kidney disease. Eur J Clin Invest 2021; 51:e13588. [PMID: 33948936 PMCID: PMC8459296 DOI: 10.1111/eci.13588] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023]
Abstract
Deregulations in gut microbiota may play a role in vascular and bone disease in chronic kidney disease (CKD). As glomerular filtration rate declines, the colon becomes more important as a site of excretion of urea and uric acid, and an increased bacterial proteolytic fermentation alters the gut microbial balance. A diet with limited amounts of fibre, as well as certain medications (eg phosphate binders, iron supplementation, antibiotics) further contribute to changes in gut microbiota composition among CKD patients. At the same time, both vascular calcification and bone disease are common in patients with advanced kidney disease. This narrative review describes emerging evidence on gut dysbiosis, vascular calcification, bone demineralization and their interrelationship termed the 'gut-bone-vascular axis' in progressive CKD. The role of diet, gut microbial metabolites (ie indoxyl sulphate, p-cresyl sulphate, trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFA)), vitamin K deficiency, inflammatory cytokines and their impact on both bone health and vascular calcification are discussed. This framework may open up novel preventive and therapeutic approaches targeting the microbiome in an attempt to improve cardiovascular and bone health in CKD.
Collapse
Affiliation(s)
- Fernanda G Rodrigues
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Nutrition Post-Graduation Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Milene S Ormanji
- Nephrology Division, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ita P Heilberg
- Nutrition Post-Graduation Program, Universidade Federal de São Paulo, São Paulo, Brazil.,Nephrology Division, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin H de Borst
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
37
|
Relationship between Circulating Galectin-3, Systemic Inflammation, and Protein-Energy Wasting in Chronic Hemodialysis Patients. Nutrients 2021; 13:nu13082803. [PMID: 34444962 PMCID: PMC8398098 DOI: 10.3390/nu13082803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 02/07/2023] Open
Abstract
Galectin-3 reportedly participates in the inflammatory process that causes insulin resistance in the target tissues. However, the role of high plasma galectin-3 levels as an indicator of protein-energy wasting (PEW) in patients undergoing maintenance hemodialysis remains unclear. This study included 240 hemodialysis patients (64.5 [55.3−74.0] years, 35.8% women) from a tertiary medical center. A baseline assessment of demographic and clinical data, biochemical parameters, and body composition was conducted. Plasma galectin-3 and other biomarkers were measured using a multiplex bead-based immunoassay. Participants were then divided into two subgroups depending on the median value of plasma galectin-3. Malnutrition was identified using the geriatric nutritional risk index (GNRI) and the criteria of the International Society of Renal Nutrition and Metabolism. Independent risk factors for elevated plasma galectin-3 and malnutrition were identified by multivariate logistic regression. The high galectin-3 group was more likely to be older, have lower lean tissue mass and GNRI scores, be diagnosed with PEW, dialyze through a tunneled catheter, and have higher circulating IL-6, TNF-α, and MCP-1 concentrations than the low galectin-3 group. After multivariate adjustment, only low mean arterial pressure, dialyzing with tunneled cuffed catheters, and elevated systemic inflammatory markers correlated with high galectin-3 levels. Plasma galectin-3 concentrations also increased significantly in hemodialysis patients with PEW. However, compared with other commonly used nutritional indicators, galectin-3 did not show superiority in predicting PEW. Although the plasma galectin-3 levels correlated with PEW severity, this correlation disappeared after adjustment for potential confounding variables (OR, 1.000; 95% CI, 0.999–1.001). In conclusion, plasma galectin-3 is a valuable biomarker for systemic inflammation but is less prominent for PEW in patients with maintenance hemodialysis. Further identification of novel biomarkers is required to detect patients at risk for malnutrition and implement appropriate interventions.
Collapse
|
38
|
Johnson SD, Olwenyi OA, Bhyravbhatla N, Thurman M, Pandey K, Klug EA, Johnston M, Dyavar SR, Acharya A, Podany AT, Fletcher CV, Mohan M, Singh K, Byrareddy SN. Therapeutic implications of SARS-CoV-2 dysregulation of the gut-brain-lung axis. World J Gastroenterol 2021; 27:4763-4783. [PMID: 34447225 PMCID: PMC8371510 DOI: 10.3748/wjg.v27.i29.4763] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
The emergence and rapid spread of novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused over 180 million confirmed cases resulting in over 4 million deaths worldwide with no clear end in sight for the coronavirus disease 19 (COVID-19) pandemic. Most SARS-CoV-2 exposed individuals experience mild to moderate symptoms, including fever, cough, fatigue, and loss of smell and taste. However, many individuals develop pneumonia, acute respiratory distress syndrome, septic shock, and multiorgan dysfunction. In addition to these primarily respiratory symptoms, SARS-CoV-2 can also infiltrate the central nervous system, which may damage the blood-brain barrier and the neuron's synapses. Resultant inflammation and neurodegeneration in the brain stem can further prevent efferent signaling to cranial nerves, leading to the loss of anti-inflammatory signaling and normal respiratory and gastrointestinal functions. Additionally, SARS-CoV-2 can infect enterocytes resulting in gut damage followed by microbial dysbiosis and translocation of bacteria and their byproducts across the damaged epithelial barrier. As a result, this exacerbates pro-inflammatory responses both locally and systemically, resulting in impaired clinical outcomes. Recent evidence has highlighted the complex interactions that mutually modulate respiratory, neurological, and gastrointestinal function. In this review, we discuss the ways SARS-CoV-2 potentially disrupts the gut-brain-lung axis. We further highlight targeting specific responses to SARS-CoV-2 for the development of novel, urgently needed therapeutic interventions. Finally, we propose a prospective related to the individuals from Low- and Middle-Income countries. Here, the underlying propensity for heightened gut damage/microbial translocation is likely to result in worse clinical outcomes during this COVID-19 pandemic.
Collapse
Affiliation(s)
- Samuel D Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Omalla A Olwenyi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Namita Bhyravbhatla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Michellie Thurman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Kabita Pandey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Elizabeth A Klug
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Morgan Johnston
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Shetty Ravi Dyavar
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, United States
| | - Kamal Singh
- Department of Molecular Microbiology and Immunology and Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, United States
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
39
|
Chai L, Luo Q, Cai K, Wang K, Xu B. Reduced fecal short-chain fatty acids levels and the relationship with gut microbiota in IgA nephropathy. BMC Nephrol 2021; 22:209. [PMID: 34082732 PMCID: PMC8173972 DOI: 10.1186/s12882-021-02414-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND IgA nephropathy(IgAN)) is the common pathological type of glomerular diseases. The role of gut microbiota in mediating "gut-IgA nephropathy" has not received sufficient attention in the previous studies. The purpose of this study was to investigate the changes of fecal short-chain fatty acids(SCFAs), a metabolite of the intestinal microbiota, in patients with IgAN and its correlation with intestinal flora and clinical indicators, and to further investigate the role of the gut-renal axis in IgAN. METHODS There were 29 patients with IgAN and 29 normal control subjects recruited from January 2018 to May 2018. The fresh feces were collected. The fecal SCFAs were measured by gas chromatography/mass spectrometry and gut microbiota was analysed by16S rDNA sequences, followed by estimation of α- and β-diversity. Correlation analysis was performed using the spearman's correlation test between SCFAs and gut microbiota. RESULTS The levels of acetic acid, propionic acid, butyric acid, isobutyric acid and caproic acid in the IgAN patients were significantly reduced compared with control group(P < 0.05). Butyric acid(r=-0.336, P = 0.010) and isobutyric acid(r=-0.298, P = 0.022) were negatively correlated with urea acid; butyric acid(r=-0.316, P = 0.016) was negatively correlated with urea nitrogen; caproic acid(r=-0.415,P = 0.025) showed negative correlation with 24-h urine protein level.Exemplified by the results of α-diversity and β-diversity, the intestinal flora of IgAN patients was significantly different from that of the control group. Acetic acid was positively associated with c_Clostridia(r = 0.357, P = 0.008), o_Clostridiales(r = 0.357, P = 0.008) and g_Eubacterium_coprostanoligenes_group(r = 0.283, P = 0.036). Butyric acid was positively associated with g_Alistipes (r = 0.278, P = 0.040). The relative abundance of those were significantly decreased in IgAN group compared to control group. CONCLUSIONS The levels of fecal SCFAs in the IgAN patients were reduced, and correlated with clinical parameters and gut microbiota, which may be involved in the pathogenesis of IgAN, and this finding may provide a new therapeutic approach.
Collapse
Affiliation(s)
- Lingxiong Chai
- Deparment of Nephrology, Ningbo Hwamei Hospital, University of Chinese Academy of Sciences, No.41, Xibei street, Zhejiang Province, 315010, Ningbo, China
- Life and Health Industry Research Institute, 315010, Ningbo, Zhejiang Province, China
| | - Qun Luo
- Deparment of Nephrology, Ningbo Hwamei Hospital, University of Chinese Academy of Sciences, No.41, Xibei street, Zhejiang Province, 315010, Ningbo, China
- Life and Health Industry Research Institute, 315010, Ningbo, Zhejiang Province, China
| | - Kedan Cai
- Deparment of Nephrology, Ningbo Hwamei Hospital, University of Chinese Academy of Sciences, No.41, Xibei street, Zhejiang Province, 315010, Ningbo, China
- Life and Health Industry Research Institute, 315010, Ningbo, Zhejiang Province, China
| | - Kaiyue Wang
- Deparment of Nephrology, Ningbo Hwamei Hospital, University of Chinese Academy of Sciences, No.41, Xibei street, Zhejiang Province, 315010, Ningbo, China
- Life and Health Industry Research Institute, 315010, Ningbo, Zhejiang Province, China
| | - Binbin Xu
- Deparment of Nephrology, Ningbo Hwamei Hospital, University of Chinese Academy of Sciences, No.41, Xibei street, Zhejiang Province, 315010, Ningbo, China.
- Life and Health Industry Research Institute, 315010, Ningbo, Zhejiang Province, China.
| |
Collapse
|
40
|
Multi-Solvent Extraction Procedure for the Pioneer Fecal Metabolomic Analysis-Identification of Potential Biomarkers in Stable Kidney Transplant Patients. Diagnostics (Basel) 2021; 11:diagnostics11060962. [PMID: 34073647 PMCID: PMC8229050 DOI: 10.3390/diagnostics11060962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/13/2021] [Accepted: 05/22/2021] [Indexed: 11/21/2022] Open
Abstract
Metabolic alteration plays a functional role in kidney allograft complications. Metabolomics is a promising high-throughput approach in nephrology but is still limited by the lack of overlap in metabolite coverage. We performed an untargeted fecal metabolomic analysis of forty stable kidney allograft recipients and twenty non-transplant controls. First, we applied the ultra-high performance liquid chromatography (UHPLC) analysis coupled with the Diod Array detector. The potential biomarkers were then collected and identified by gas chromatography-mass spectrometry (GCMS). In order to allow for complete coverage of the fecal polar and non-polar metabolites, the performance of five organic solvents with increasing polarity was investigated successively. UHPLC analysis revealed that the fecal metabolite profiles following the five extractions were significantly different between controls and kidney allografts. GC-MS analysis showed that the best predictors’ metabolites belonged mainly to long-chain fatty acids, phenolic compounds, and amino acids. Collectively, our results showed the efficiency of our pioneer method to successfully discriminate stable kidney-transplant recipients from controls. These findings suggest that distinct metabolic profiles mainly affect fatty acid biosynthesis and amino acid metabolism. In such a context, the novel insights into metabolomic investigation may be a valuable tool that could provide useful new relevant biomarkers for preventing kidney transplant complications.
Collapse
|
41
|
Luo D, Zhao W, Lin Z, Wu J, Lin H, Li Y, Song J, Zhang J, Peng H. The Effects of Hemodialysis and Peritoneal Dialysis on the Gut Microbiota of End-Stage Renal Disease Patients, and the Relationship Between Gut Microbiota and Patient Prognoses. Front Cell Infect Microbiol 2021; 11:579386. [PMID: 33834002 PMCID: PMC8021868 DOI: 10.3389/fcimb.2021.579386] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/25/2021] [Indexed: 11/15/2022] Open
Abstract
Gut microbiota alterations occur in end-stage renal disease (ESRD) patients with or without dialysis. However, it remains unclear whether changes in gut microbiota of dialysis ESRD patients result from dialysis or ESRD, or both. Similarly, there is a dearth of information on the relationship between gut microbiota and ESRD prognoses. We collected fecal samples and tracked clinical outcomes from 73 ESRD patients, including 33 pre-dialysis ESRD patients, 19 peritoneal dialysis (PD) patients, and 21 hemodialysis (HD) patients. 16S rRNA sequencing and bioinformatics tools were used to analyze the gut microbiota of ESRD patients and healthy controls. Gut microbiota diversity was different before and after dialysis. Bacteroidetes were significantly deceased in HD patients. Twelve bacterial genera exhibited statistically significant differences, due to dialysis (all P < 0.05, FDR corrected). HD reversed abnormal changes in Oscillospira and SMB53 in pre-dialysis patients. Functional predictions of microbial communities showed that PD and HD altered signal transduction and metabolic pathways in ESRD patients. Furthermore, Bacteroides and Phascolarctobacterium were associated with cardiovascular mortality. Dorea, Clostridium, and SMB53 were related to peritonitis in PD patients. This study not only demonstrated differences in gut microbiota between pre-dialysis and dialysis ESRD patients, but also firstly proposed gut bacteria may exert an impact on patient prognosis.
Collapse
Affiliation(s)
- Dan Luo
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenbo Zhao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiming Lin
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianhao Wu
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongchun Lin
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yongjie Li
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Song
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Zhang
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
Dong S, Zeng B, Hu L, Zhang Y, Xiong J, Deng J, Huang L, Liao Z, Wang J, Wei H, Fang X. Effect of a Humanized Diet Profile on Colonization Efficiency and Gut Microbial Diversity in Human Flora-Associated Mice. Front Nutr 2021; 8:633738. [PMID: 33708788 PMCID: PMC7940529 DOI: 10.3389/fnut.2021.633738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Human flora-associated (HFA) mouse models allow us to design interventions for human disease research to test specific hypotheses and explore the complex commensal microbiome while avoiding the ethical limitations of using humans as models to directly study intestinal flora diseases. However, few studies have investigated the effect of a humanized diet profile (coarse-feed diet; CFD) on colonization efficiency and gut microbial diversity in HFA mice. We tested the colonization efficiency and gut microbial diversity in germ-free Kunming (KM) mice fed a CFD or a purified feed diet (PFD) at 1, 2, and 4 weeks. Although the colonization efficiencies differed significantly (67.50–70.00% vs. 72.69–85.96%) in the HFA mice, the colonization efficiency of the PFD-fed HFA mice (85.96%) was significantly higher than that of the CFD-fed mice (69.61%) at 2 weeks. At 4 weeks, the colonization efficiency of the PFD-fed mice (72.69%) was comparable to that of the CFD-fed mice (70.00%). Additionally, the gut microbial diversity of the CFD-fed HFA mice was similar to that of a human fecal donor. Regarding the Kyoto Encyclopedia of Genes and Genomes colonic microbiota metabolic pathways, the CFD-fed HFA mice showed more similarities to the human donor than to the PFD-fed mice in amino sugar and nucleotide sugar metabolism, biosynthesis of amino acids, carbon metabolism, purine metabolism, and phosphotransferase systems. In conclusion, the humanized diet profiles of the CFD and PFD could help establish human microbiotas in mice. Constructing HFA mouse models fed a CFD for 4 weeks may be useful in researching human-derived intestinal diseases.
Collapse
Affiliation(s)
- Sashuang Dong
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - BenHua Zeng
- Department of Laboratory Animal Science, College of Basic Medicine Science, Third Military Medical University, Chongqing, China
| | - Ling Hu
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Yuling Zhang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jiaqi Xiong
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jing Deng
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Liyan Huang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - ZhenLin Liao
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jie Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medicine Science, Third Military Medical University, Chongqing, China.,Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Fang
- College of Food Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
43
|
McCarthy CG, Saha P, Golonka RM, Wenceslau CF, Joe B, Vijay-Kumar M. Innate Immune Cells and Hypertension: Neutrophils and Neutrophil Extracellular Traps (NETs). Compr Physiol 2021; 11:1575-1589. [PMID: 33577121 DOI: 10.1002/cphy.c200020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Uncontrolled immune system activation amplifies end-organ injury in hypertension. Nonetheless, the exact mechanisms initiating this exacerbated inflammatory response, thereby contributing to further increases in blood pressure (BP), are still being revealed. While participation of lymphoid-derived immune cells has been well described in the hypertension literature, the mechanisms by which myeloid-derived innate immune cells contribute to T cell activation, and subsequent BP elevation, remains an active area of investigation. In this article, we critically analyze the literature to understand how monocytes, macrophages, dendritic cells, and polymorphonuclear leukocytes, including mast cells, eosinophils, basophils, and neutrophils, contribute to hypertension and hypertension-associated end-organ injury. The most abundant leukocytes, neutrophils, are indisputably increased in hypertension. However, it is unknown how (and why) they switch from critical first responders of the innate immune system, and homeostatic regulators of BP, to tissue-damaging, pro-hypertensive mediators. We propose that myeloperoxidase-derived pro-oxidants, neutrophil elastase, neutrophil extracellular traps (NETs), and interactions with other innate and adaptive immune cells are novel mechanisms that could contribute to the inflammatory cascade in hypertension. We further posit that the gut microbiota serves as a set point for neutropoiesis and their function. Finally, given that hypertension appears to be a key risk factor for morbidity and mortality in COVID-19 patients, we put forth evidence that neutrophils and NETs cause cardiovascular injury post-coronavirus infection, and thus may be proposed as an intriguing therapeutic target for high-risk individuals. © 2021 American Physiological Society. Compr Physiol 11:1575-1589, 2021.
Collapse
Affiliation(s)
- Cameron G McCarthy
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Piu Saha
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Rachel M Golonka
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Camilla F Wenceslau
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Bina Joe
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Matam Vijay-Kumar
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
44
|
Lai L, Li Y, Liu J, Luo L, Tang J, Xue J, Liu T. Bovine serum albumin aggravates macrophage M1 activation and kidney injury in heterozygous Klotho-deficient mice via the gut microbiota-immune axis. Int J Biol Sci 2021; 17:742-755. [PMID: 33767585 PMCID: PMC7975693 DOI: 10.7150/ijbs.56424] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Klotho expression abnormalities induces kidney injury and chronic kidney disease, however, the underlying mechanism remains unclear. Here, Klotho+/- mice and wild-type mice were treated with low-dose bovine serum albumin (BSA). Pathological examination demonstrated that the area of glomerular collagen deposition and fibrosis in BSA-Kl-/+ mice was significantly larger than that in BSA-WT mice. The serum levels of superoxide dismutase, malondialdehyde, creatinine, and urea in BSA-Kl-/+ mice were significantly increased. Sequencing of gut microbiota 16S rRNA v3-v4 region indicated that BSA-Kl-/+ mice showed a significantly higher relative abundance of the genera Dubosiella, Akkermansia, Alloprevotella, and Lachnospiraceae and a significantly lower relative abundance of the genera Allobaculum and Muribaculaceae than BSA-WT mice. KEGG analysis revealed that the metabolic pathways of signal transduction, xenobiotic biodegradation and metabolism, and lipid metabolism increased significantly in BSA-Kl-/+ mice. Flow cytometry showed that the proportion of CD68+/CD11b+ cells in the peripheral blood was significantly higher in BSA-KL-/+ mice than that in BSA-WT mice. qPCR and western blot suggested that Klotho and Nrf2 expression in MΦ1 cells of BSA-KL-/+ mice was significantly decreased. Thus, the findings suggest during the immune activation and chronic inflammation induced by the gut microbiota imbalance in Klotho-deficient mice treated to BSA, disrupted expression of proteins in the Nrf2/NF-κB signaling pathway in monocyte-derived macrophage M1 cells leads to the aggravation of inflammation and kidney injury.
Collapse
Affiliation(s)
- Lingyun Lai
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yi Li
- Division of Nephrology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianjun Liu
- Trauma-Emergency & Critical Care Medicine Center, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Lei Luo
- Division of Nephrology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianguo Tang
- Trauma-Emergency & Critical Care Medicine Center, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Jun Xue
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China.,Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
45
|
Ma E, Ohira T, Yasumura S, Nakano H, Eguchi E, Miyazaki M, Hosoya M, Sakai A, Takahashi A, Ohira H, Kazama J, Shimabukuro M, Yabe H, Maeda M, Ohto H, Kamiya K. Dietary Patterns and Progression of Impaired Kidney Function in Japanese Adults: A Longitudinal Analysis for the Fukushima Health Management Survey, 2011-2015. Nutrients 2021; 13:nu13010168. [PMID: 33430501 PMCID: PMC7827845 DOI: 10.3390/nu13010168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
To investigate associations between dietary patterns and the risk of impaired kidney function, we analyzed data from 14,732 participants (40–89 years) who completed the baseline diet questionnaire of The Fukushima Health Management Survey in 2011. The incidence of chronic kidney disease (CKD) (estimated glomerular filtration rate (eGFR) <60 mL/min/1.73 m2 or proteinuria (≥1+ by dipstick test)) and annual changes in eGFR were assessed from 2012 to 2015. Three major dietary patterns were identified. The adjusted cumulative incidence ratio of the highest vs. lowest tertile of a vegetable diet scores was 0.90 (95% confidence interval (CI): 0.82, 1.00) for eGFR < 60 mL/min/1.73 m2, 0.68 (95% CI: 0.52, 0.90) for proteinuria, and 0.88 (95% CI: 0.80, 0.97) for CKD (P for trend = 0.031, 0.007, and 0.005, respectively). The incident risk of CKD in the highest tertile of juice diet scores was 18% higher than the lowest tertile. The odds ratio of the highest vs. lowest tertile of vegetable diet scores was 0.85 (95% CI: 0.75, 0.98) in the rapidly decreasing eGFR group (P for trend = 0.009). We did not observe significant associations for the meat dietary pattern. A Japanese vegetable diet could reduce the risk of developing impaired kidney function and CKD.
Collapse
Affiliation(s)
- Enbo Ma
- Health Promotion Centre, Fukushima Medical University, Fukushima 960-1295, Japan; (T.O.); (M.M.); (M.H.)
- Department of Epidemiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; (H.N.); (E.E.)
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Correspondence: ; Tel.: +81-24-547-1788; Fax: +81-24-547-1789
| | - Tetsuya Ohira
- Health Promotion Centre, Fukushima Medical University, Fukushima 960-1295, Japan; (T.O.); (M.M.); (M.H.)
- Department of Epidemiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; (H.N.); (E.E.)
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
| | - Seiji Yasumura
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Public Health, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Hironori Nakano
- Department of Epidemiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; (H.N.); (E.E.)
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
| | - Eri Eguchi
- Department of Epidemiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan; (H.N.); (E.E.)
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
| | - Makoto Miyazaki
- Health Promotion Centre, Fukushima Medical University, Fukushima 960-1295, Japan; (T.O.); (M.M.); (M.H.)
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
| | - Mitsuaki Hosoya
- Health Promotion Centre, Fukushima Medical University, Fukushima 960-1295, Japan; (T.O.); (M.M.); (M.H.)
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Paediatrician, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Akira Sakai
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Radiation Life Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Atsushi Takahashi
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan;
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan;
| | - Junichiro Kazama
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Nephrology and Hypertension, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Michio Shimabukuro
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Diabetes, Endocrinology, and Metabolism, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Hirooki Yabe
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Neuropsychiatry, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masaharu Maeda
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Department of Disaster Psychiatry, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Hitoshi Ohto
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
| | - Kenji Kamiya
- Radiation Medical Science Centre for Fukushima Health Management Survey, Fukushima Medical University, Fukushima 960-1295, Japan; (S.Y.); (A.S.); (A.T.); (J.K.); (M.S.); (H.Y.); (M.M.); (H.O.); (K.K.)
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
46
|
Hajiagha MN, Taghizadeh S, Asgharzadeh M, Dao S, Ganbarov K, Köse Ş, Kafil HS. Gut microbiota And Human Body Interactions; Its Impact on Health: a review. Curr Pharm Biotechnol 2021; 23:4-14. [PMID: 33397232 DOI: 10.2174/1389201022666210104115836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022]
Abstract
Gut microbiota (GM) as an organ of the human body has a particular and autonomous function that related to it. This review aimed to investigate human intestinal and gut microbiota interaction and its impact on health. As a creation referable database about this dynamic and complex organ, several comprehensive projects are implemented by using culture-dependent (culturomics), culture independent methods (e.g metagenomics, mathematics model), and Gnotobiological together. This study was done by searching PubMed, Scopus and Google scholar database in the gut, health microbiota and interaction keywords. The first acquired microbiota during pregnancy or childbirth is colonized in the gut by using specific and non-specific mechanisms. That`s structure and shape reach relative stability with selection pressure along with host development until adulthood and keep its resilience against external or internal variables depending on the host genetics and negative feedback. Due to several research individuals have 2 functional group microbiota including the core (common between vast majorities human) and flexible (transient population) microbiome. The most important role of the GM in the human body can be summarized in three basic landscapes: metabolic, immune system, and gut-brain axis interaction. So that loss of microbial population balance will lead to disorder and disease.
Collapse
Affiliation(s)
| | - Sepehr Taghizadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Mohammad Asgharzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Sounkalo Dao
- Faculté de Médecine, de Pharmacie et d'Odonto-Stomatologie (FMPOS), University of Bamako, Bamako. Mali
| | | | - Şükran Köse
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, İzmir. Turkey
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
47
|
Gabr M, Świderek K. Discovery of a Histidine-Based Scaffold as an Inhibitor of Gut Microbial Choline Trimethylamine-Lyase. ChemMedChem 2020; 15:2273-2279. [PMID: 32827245 DOI: 10.1002/cmdc.202000571] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Indexed: 01/07/2023]
Abstract
Anaerobic choline metabolism by human gut microbiota to produce trimethylamine (TMA) has recently evolved as a potential therapeutic target because of its association with chronic kidney disease and increased cardiovascular risks. Limited examples of choline analogues have been reported as inhibitors of bacterial enzyme choline TMA-lyase (CutC), a key enzyme regulating choline anaerobic metabolism. We used a new workflow to discover CutC inhibitors based on focused screening of a diversified library of small molecules for intestinal metabolic stability followed by in vitro CutC inhibitory assay. This workflow identified a histidine-based scaffold as a CutC inhibitor with an IC50 value of 1.9±0.2 μM. Remarkably, the identified CutC inhibitor was able to reduce the production of TMA in whole-cell assays using various bacterial strains as well as in complex gut microbiota environment. The improved efficiency of the new scaffold identified in this study in comparison to previously reported CutC inhibitors would enable optimization of potential leads for in vivo screening and clinical translation. Finally, docking studies and molecular-dynamic simulations were used to predict putative interactions created between inhibitor and CutC.
Collapse
Affiliation(s)
- Moustafa Gabr
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Katarzyna Świderek
- Departament de Química Física i Analítica, Universitat Jaume I, 12071, Castellón, Spain
| |
Collapse
|
48
|
Bakkar NMZ, Dwaib HS, Fares S, Eid AH, Al-Dhaheri Y, El-Yazbi AF. Cardiac Autonomic Neuropathy: A Progressive Consequence of Chronic Low-Grade Inflammation in Type 2 Diabetes and Related Metabolic Disorders. Int J Mol Sci 2020; 21:E9005. [PMID: 33260799 PMCID: PMC7730941 DOI: 10.3390/ijms21239005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac autonomic neuropathy (CAN) is one of the earliest complications of type 2 diabetes (T2D), presenting a silent cause of cardiovascular morbidity and mortality. Recent research relates the pathogenesis of cardiovascular disease in T2D to an ensuing chronic, low-grade proinflammatory and pro-oxidative environment, being the hallmark of the metabolic syndrome. Metabolic inflammation emerges as adipose tissue inflammatory changes extending systemically, on the advent of hyperglycemia, to reach central regions of the brain. In light of changes in glucose and insulin homeostasis, dysbiosis or alteration of the gut microbiome (GM) emerges, further contributing to inflammatory processes through increased gut and blood-brain barrier permeability. Interestingly, studies reveal that the determinants of oxidative stress and inflammation progression exist at the crossroad of CAN manifestations, dictating their evolution along the natural course of T2D development. Indeed, sympathetic and parasympathetic deterioration was shown to correlate with markers of adipose, vascular, and systemic inflammation. Additionally, evidence points out that dysbiosis could promote a sympatho-excitatory state through differentially affecting the secretion of hormones and neuromodulators, such as norepinephrine, serotonin, and γ-aminobutyric acid, and acting along the renin-angiotensin-aldosterone axis. Emerging neuronal inflammation and concomitant autophagic defects in brainstem nuclei were described as possible underlying mechanisms of CAN in experimental models of metabolic syndrome and T2D. Drugs with anti-inflammatory characteristics provide potential avenues for targeting pathways involved in CAN initiation and progression. The aim of this review is to delineate the etiology of CAN in the context of a metabolic disorder characterized by elevated oxidative and inflammatory load.
Collapse
Affiliation(s)
- Nour-Mounira Z. Bakkar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh 1107 2020, Beirut 11-0236, Lebanon; (N.-M.Z.B.); (H.S.D.); (A.H.E.)
| | - Haneen S. Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh 1107 2020, Beirut 11-0236, Lebanon; (N.-M.Z.B.); (H.S.D.); (A.H.E.)
| | - Souha Fares
- Rafic Hariri School of Nursing, American University of Beirut, Riad El-Solh 1107 2020, Beirut 11-0236, Lebanon;
| | - Ali H. Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh 1107 2020, Beirut 11-0236, Lebanon; (N.-M.Z.B.); (H.S.D.); (A.H.E.)
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Yusra Al-Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, UAE
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh 1107 2020, Beirut 11-0236, Lebanon; (N.-M.Z.B.); (H.S.D.); (A.H.E.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
49
|
Souai N, Zidi O, Mosbah A, Kosai I, Manaa JE, Mokhtar NB, Asimakis E, Stathopoulou P, Cherif A, Tsiamis G, Kouidhi S. Impact of the Post-Transplant Period and Lifestyle Diseases on Human Gut Microbiota in Kidney Graft Recipients. Microorganisms 2020; 8:microorganisms8111724. [PMID: 33158078 PMCID: PMC7694191 DOI: 10.3390/microorganisms8111724] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
Gaining long-term graft function and patient life quality remain critical challenges following kidney transplantation. Advances in immunology, gnotobiotics, and culture-independent molecular techniques have provided growing insights into the complex relationship of the microbiome and the host. However, little is known about the over time-shift of the gut microbiota in the context of kidney transplantation and its impact on both graft and health stability. Here we aimed to characterize the structure of gut microbiota within stable kidney graft recipients. We enrolled forty kidney transplant patients after at least three months of transplantation and compared them to eighteen healthy controls. The overall microbial community structure of the kidney transplanted group was clearly different from control subjects. We found lower relative abundances of Actinobacteria, Bacteroidetes, and Verrucomicrobia within the patient group and a higher abundance of Proteobacteria compared to the control group. Both richness and Shannon diversity indexes were significantly lower in the kidney graft recipients than in healthy controls. Post-graft period was positively correlated with the relative abundance of the Proteobacteria phylum, especially Escherichia.Shigella genus. Interestingly, only Parabacteroides was found to significantly differentiate patients that were not suffering from lifestyle diseases and those who suffer from post-graft complications. Furthermore, network analysis showed that the occurrence of lifestyle diseases was significantly linked with a higher number of negative interactions of Sutterella and Succinivibrio genera within patients. This study characterizes gut microbiome fluctuation in stable kidney transplant patients after a long post-allograft period. Analysis of fecal microbiota could be useful for nephrologists as a new clinical tool that can improve kidney allograft monitoring and outcomes.
Collapse
Affiliation(s)
- Nessrine Souai
- Laboratory of Biotechnology and Valorisation of Bio-GeoRessources, Higher Institute of Biotechnology of Sidi Thabet, BiotechPole of Sidi Thabet, University of Manouba, Ariana 2020, Tunisia; (N.S.); (O.Z.); (A.M.); (A.C.)
- Department of Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Farhat Hachad Universitary Campus, Rommana 1068, Tunis, Tunisia
| | - Oumaima Zidi
- Laboratory of Biotechnology and Valorisation of Bio-GeoRessources, Higher Institute of Biotechnology of Sidi Thabet, BiotechPole of Sidi Thabet, University of Manouba, Ariana 2020, Tunisia; (N.S.); (O.Z.); (A.M.); (A.C.)
- Department of Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Farhat Hachad Universitary Campus, Rommana 1068, Tunis, Tunisia
| | - Amor Mosbah
- Laboratory of Biotechnology and Valorisation of Bio-GeoRessources, Higher Institute of Biotechnology of Sidi Thabet, BiotechPole of Sidi Thabet, University of Manouba, Ariana 2020, Tunisia; (N.S.); (O.Z.); (A.M.); (A.C.)
| | - Imen Kosai
- Unit of Organ Transplant Military Training Hospital, Mont Fleury 1008, Tunis, Tunisia; (I.K.); (J.E.M.)
| | - Jameleddine El Manaa
- Unit of Organ Transplant Military Training Hospital, Mont Fleury 1008, Tunis, Tunisia; (I.K.); (J.E.M.)
| | - Naima Bel Mokhtar
- Laboratory of Systems Microbiology and Applied Genomics, Department of Environmental Engineering, University of Patras, 2 Seferi St, 30100 Agrinio, Greece; (N.B.M.); (E.A.); (P.S.); (G.T.)
| | - Elias Asimakis
- Laboratory of Systems Microbiology and Applied Genomics, Department of Environmental Engineering, University of Patras, 2 Seferi St, 30100 Agrinio, Greece; (N.B.M.); (E.A.); (P.S.); (G.T.)
| | - Panagiota Stathopoulou
- Laboratory of Systems Microbiology and Applied Genomics, Department of Environmental Engineering, University of Patras, 2 Seferi St, 30100 Agrinio, Greece; (N.B.M.); (E.A.); (P.S.); (G.T.)
| | - Ameur Cherif
- Laboratory of Biotechnology and Valorisation of Bio-GeoRessources, Higher Institute of Biotechnology of Sidi Thabet, BiotechPole of Sidi Thabet, University of Manouba, Ariana 2020, Tunisia; (N.S.); (O.Z.); (A.M.); (A.C.)
| | - George Tsiamis
- Laboratory of Systems Microbiology and Applied Genomics, Department of Environmental Engineering, University of Patras, 2 Seferi St, 30100 Agrinio, Greece; (N.B.M.); (E.A.); (P.S.); (G.T.)
| | - Soumaya Kouidhi
- Laboratory of Biotechnology and Valorisation of Bio-GeoRessources, Higher Institute of Biotechnology of Sidi Thabet, BiotechPole of Sidi Thabet, University of Manouba, Ariana 2020, Tunisia; (N.S.); (O.Z.); (A.M.); (A.C.)
- Correspondence: ; Tel.: +216-95-694-135
| |
Collapse
|
50
|
Kim SM, Song IH. The clinical impact of gut microbiota in chronic kidney disease. Korean J Intern Med 2020; 35:1305-1316. [PMID: 32872729 PMCID: PMC7652652 DOI: 10.3904/kjim.2020.411] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
Gut microorganisms play critical roles in both maintaining host homeostasis and the development of diverse diseases. Gut dysbiosis, an alteration of the composition and function of gut microorganisms, is commonly seen in patients with chronic kidney disease (CKD). CKD itself contributes to a disruption of the symbiotic relationship between the gut microbiota and the host, while the resulting gut dysbiosis may play a part in stage progression of CKD. This bidirectional relationship supports the concept that the gut microbiota is considered a novel focus for the pathogenesis and management of CKD. This article examines the interaction between the gut microbiota and the kidney, the mutual effects of dysbiosis and CKD, and possible treatment options to restore gut eubiosis, and reduce CKD progression and its related complications.
Collapse
Affiliation(s)
- So Mi Kim
- Division of Nephrology, Department of Internal Medicine, Dankook University Hospital, Cheonan, Korea
| | - Il han Song
- Division of Hepatology, Department of Internal Medicine, Dankook University Hospital, Cheonan, Korea
- Correspondence to Il Han Song, M.D. Division of Hepatology, Department of Internal Medicine, Dankook University Hospital, 201 Manghyang-ro, Dongnam-gu, Cheonan 31116, Korea Tel: +82-41-550-3924 Fax: +82-41-556-3256 E-mail:
| |
Collapse
|