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
|
Yafarova AA, Dementeva EV, Zlobovskaya OA, Sheptulina AF, Lopatukhina EV, Timofeev YS, Glazunova EV, Lyundup AV, Doludin YV, Kiselev AR, Shipulin GA, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota and Metabolic Alterations Associated with Heart Failure and Coronary Artery Disease. Int J Mol Sci 2024; 25:11295. [PMID: 39457077 PMCID: PMC11508380 DOI: 10.3390/ijms252011295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced ejection fraction (HFrEF). This cross-sectional study included 189 participants divided into three groups: coronary artery disease (n = 93), heart failure with reduced ejection fraction (n = 43), and controls (n = 53). Assessments included physical exams, echocardiography, dietary surveys, blood analysis, and fecal analysis. Gut microbiota composition was analyzed using next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR). Statistical analysis methods for testing hypotheses and correlations, alpha and beta-diversity analyses, co-occurrence networks, and machine learning were conducted using Python libraries or R packages with multiple comparisons corrected using the Benjamini-Hochberg procedure. Significant gut microbiota alterations were observed, with higher Bacillota/Bacteroidota ratios in CAD and HFrEF groups compared to controls (p < 0.001). Significant differences were observed in α-diversity indices (Pielou, Chao1, Faith) between disease groups and controls (p < 0.001). β-diversity analyses also revealed distinct microbial profiles (p = 0.0015). Interestingly, trimethylamine N-oxide (TMAO) levels were lower in CAD and HFrEF groups compared to controls (p < 0.05), while indoxyl sulfate (IS) levels were comparable between the study groups. Co-occurrence network analysis and machine learning identified key microbial features linked to these conditions, highlighting complex interactions within the gut microbiota associated with cardiovascular disease.
Collapse
Affiliation(s)
- Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Dementeva
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Olga A. Zlobovskaya
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Lopatukhina
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Yuriy S. Timofeev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Evgeniya V. Glazunova
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Aleksey V. Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, 117036 Moscow, Russia
| | - Yuriy V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - German A. Shipulin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Valentin V. Makarov
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Sergey M. Yudin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| |
Collapse
|
3
|
Zhao X, Gao J, Kou K, Wang X, Gao X, Wang Y, Zhou H, Li F. Causal effects of plasma metabolites on chronic kidney diseases and renal function: a bidirectional Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1429159. [PMID: 39129920 PMCID: PMC11310041 DOI: 10.3389/fendo.2024.1429159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Background Despite the potential demonstrated by targeted plasma metabolite modulators in halting the progression of chronic kidney disease (CKD), a lingering uncertainty persists concerning the causal relationship between distinct plasma metabolites and the onset and progression of CKD. Methods A genome-wide association study was conducted on 1,091 metabolites and 309 metabolite ratios derived from a cohort of 8,299 unrelated individuals of European descent. Employing a bidirectional two-sample Mendelian randomization (MR) analysis in conjunction with colocalization analysis, we systematically investigated the associations between these metabolites and three phenotypes: CKD, creatinine-estimated glomerular filtration rate (creatinine-eGFR), and urine albumin creatinine ratio (UACR). In the MR analysis, the primary analytical approach employed was inverse variance weighting (IVW), and sensitivity analysis was executed utilizing the MR-Egger method and MR-pleiotropy residual sum and outlier (MR-PRESSO). Heterogeneity was carefully evaluated through Cochrane's Q test. To ensure the robustness of our MR results, the leave-one-out method was implemented, and the strength of causal relationships was subjected to scrutiny via Bonferroni correction. Results Our thorough MR analysis involving 1,400 plasma metabolites and three clinical phenotypes yielded a discerning identification of 21 plasma metabolites significantly associated with diverse outcomes. Specifically, in the forward MR analysis, 6 plasma metabolites were determined to be causally associated with CKD, 16 with creatinine-eGFR, and 7 with UACR. Substantiated by robust evidence from colocalization analysis, 6 plasma metabolites shared causal variants with CKD, 16 with creatinine-eGFR, and 7 with UACR. In the reverse analysis, a diminished creatinine-eGFR was linked to elevated levels of nine plasma metabolites. Notably, no discernible associations were observed between other plasma metabolites and CKD, creatinine-eGFR, and UACR. Importantly, our analysis detected no evidence of horizontal pleiotropy. Conclusion This study elucidates specific plasma metabolites causally associated with CKD and renal functions, providing potential targets for intervention. These findings contribute to an enriched understanding of the genetic underpinnings of CKD and renal functions, paving the way for precision medicine applications and therapeutic strategies aimed at impeding disease progression.
Collapse
Affiliation(s)
- Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Jialin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Kai Kou
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xi Wang
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Tian K, Jing D, Lan J, Lv M, Wang T. Commensal microbiome and gastrointestinal mucosal immunity: Harmony and conflict with our closest neighbor. Immun Inflamm Dis 2024; 12:e1316. [PMID: 39023417 PMCID: PMC11256888 DOI: 10.1002/iid3.1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND The gastrointestinal tract contains a wide range of microorganisms that have evolved alongside the immune system of the host. The intestinal mucosa maintains balance within the intestines by utilizing the mucosal immune system, which is controlled by the complex gut mucosal immune network. OBJECTIVE This review aims to comprehensively introduce current knowledge of the gut mucosal immune system, focusing on its interaction with commensal bacteria. RESULTS The gut mucosal immune network includes gut-associated lymphoid tissue, mucosal immune cells, cytokines, and chemokines. The connection between microbiota and the immune system occurs through the engagement of bacterial components with pattern recognition receptors found in the intestinal epithelium and antigen-presenting cells. This interaction leads to the activation of both innate and adaptive immune responses. The interaction between the microbial community and the host is vital for maintaining the balance and health of the host's mucosal system. CONCLUSION The gut mucosal immune network maintains a delicate equilibrium between active immunity, which defends against infections and damaging non-self antigens, and immunological tolerance, which allows for the presence of commensal microbiota and dietary antigens. This balance is crucial for the maintenance of intestinal health and homeostasis. Disturbance of gut homeostasis leads to enduring or severe gastrointestinal ailments, such as colorectal cancer and inflammatory bowel disease. Utilizing these factors can aid in the development of cutting-edge mucosal vaccines that have the ability to elicit strong protective immune responses at the primary sites of pathogen invasion.
Collapse
Affiliation(s)
- Kexin Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Dehong Jing
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Junzhe Lan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Mingming Lv
- Department of BreastWomen's Hospital of Nanjing Medical University, Nanjing Maternity, and Child Health Care HospitalNanjingChina
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
5
|
Liu M, He P, Ye Z, Yang S, Zhang Y, Wu Q, Zhou C, Zhang Y, Hou FF, Qin X. Functional gastrointestinal disorders, mental health, genetic susceptibility, and incident chronic kidney disease. Chin Med J (Engl) 2024; 137:1088-1094. [PMID: 37668042 PMCID: PMC11062687 DOI: 10.1097/cm9.0000000000002805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Whether functional gastrointestinal disorders (FGIDs) are associated with the long-term risk of chronic kidney disease (CKD) remains unclear. We aimed to investigate the prospective association of FGIDs with CKD and examine whether mental health mediated the association. METHODS About 416,258 participants without a prior CKD diagnosis enrolled in the UK Biobank between 2006 and 2010 were included. Participants with FGIDs (including irritable bowel syndrome [IBS], dyspepsia, and other functional intestinal disorders [FIDs; mainly composed of constipation]) were the exposure group, and non-FGID participants were the non-exposure group. The primary outcome was incident CKD, ascertained from hospital admission and death registry records. A Cox proportional hazard regression model was used to investigate the association between FGIDs and CKD, and the mediation analysis was performed to investigate the mediation proportions of mental health. RESULTS At baseline, 33,156 (8.0%) participants were diagnosed with FGIDs, including 21,060 (5.1%), 8262 (2.0%), and 6437 (1.6%) cases of IBS, dyspepsia, and other FIDs, respectively. During a mean follow-up period of 12.1 years, 11,001 (2.6%) participants developed CKD. FGIDs were significantly associated with a higher risk of incident CKD compared to the absence of FGIDs (hazard ratio [HR], 1.36; 95% confidence interval [CI], 1.28-1.44). Similar results were observed for IBS (HR, 1.27; 95% CI, 1.17-1.38), dyspepsia (HR, 1.30; 95% CI, 1.17-1.44), and other FIDs (HR, 1.60; 95% CI, 1.43-1.79). Mediation analyses suggested that the mental health score significantly mediated 9.05% of the association of FGIDs with incident CKD and 5.63-13.97% of the associations of FGID subtypes with CKD. Specifically, the positive associations of FGIDs and FGID subtypes with CKD were more pronounced in participants with a high genetic risk of CKD. CONCLUSION Participants with FGIDs had a higher risk of incident CKD, which was partly explained by mental health scores and was more pronounced in those with high genetic susceptibility to CKD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong 510515, China
| |
Collapse
|
6
|
Paraskevaidis I, Briasoulis A, Tsougos E. Oral Cardiac Drug-Gut Microbiota Interaction in Chronic Heart Failure Patients: An Emerging Association. Int J Mol Sci 2024; 25:1716. [PMID: 38338995 PMCID: PMC10855150 DOI: 10.3390/ijms25031716] [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: 12/05/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Regardless of the currently proposed best medical treatment for heart failure patients, the morbidity and mortality rates remain high. This is due to several reasons, including the interaction between oral cardiac drug administration and gut microbiota. The relation between drugs (especially antibiotics) and gut microbiota is well established, but it is also known that more than 24% of non-antibiotic drugs affect gut microbiota, altering the microbe's environment and its metabolic products. Heart failure treatment lies mainly in the blockage of neuro-humoral hyper-activation. There is debate as to whether the administration of heart-failure-specific drugs can totally block this hyper-activation, or whether the so-called intestinal dysbiosis that is commonly observed in this group of patients can affect their action. Although there are several reports indicating a strong relation between drug-gut microbiota interplay, little is known about this relation to oral cardiac drugs in chronic heart failure. In this review, we review the contemporary data on a topic that is in its infancy. We aim to produce scientific thoughts and questions and provide reasoning for further clinical investigation.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- Division of Cardiology, Hygeia Hospital, Erithrou Stavrou 4, 15123 Athens, Greece;
- Heart Failure Subdivision, Department of Clinical Therapeutics, Alexandra Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Vassilisis Sofias 80, 11528 Athens, Greece;
| | - Alexandros Briasoulis
- Heart Failure Subdivision, Department of Clinical Therapeutics, Alexandra Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Vassilisis Sofias 80, 11528 Athens, Greece;
| | - Elias Tsougos
- Division of Cardiology, Hygeia Hospital, Erithrou Stavrou 4, 15123 Athens, Greece;
| |
Collapse
|
7
|
Zhang J, Zhu P, Li S, Gao Y, Xing Y. From heart failure and kidney dysfunction to cardiorenal syndrome: TMAO may be a bridge. Front Pharmacol 2023; 14:1291922. [PMID: 38074146 PMCID: PMC10703173 DOI: 10.3389/fphar.2023.1291922] [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: 09/10/2023] [Accepted: 11/13/2023] [Indexed: 10/23/2024] Open
Abstract
The study of trimethylamine oxide (TMAO), a metabolite of gut microbiota, and heart failure and chronic kidney disease has made preliminary achievements and been summarized by many researchers, but its research in the field of cardiorenal syndrome is just beginning. TMAO is derived from the trimethylamine (TMA) that is produced by the gut microbiota after consumption of carnitine and choline and is then transformed by flavin-containing monooxygenase (FMO) in the liver. Numerous research results have shown that TMAO not only participates in the pathophysiological progression of heart and renal diseases but also significantly affects outcomes in chronic heart failure (CHF) and chronic kidney disease (CKD), besides influencing the general health of populations. Elevated circulating TMAO levels are associated with adverse cardiovascular events such as HF, myocardial infarction, and stroke, patients with CKD have a poor prognosis as well. However, no study has confirmed an association between TMAO and cardiorenal syndrome (CRS). As a syndrome in which heart and kidney diseases intersect, CRS is often overlooked by clinicians. Here, we summarize the research on TMAO in HF and kidney disease and review the existing biomarkers of CRS. At the same time, we introduced the relationship between exercise and gut microbiota, and appropriately explored the possible mechanisms by which exercise affects gut microbiota. Finally, we discuss whether TMAO can serve as a biomarker of CRS, with the aim of providing new strategies for the detection, prognostic, and treatment evaluation of CRS.
Collapse
Affiliation(s)
- Jialun Zhang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Peining Zhu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Siyu Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yufei Gao
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yue Xing
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Paraskevaidis I, Xanthopoulos A, Tsougos E, Triposkiadis F. Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ. Biomedicines 2023; 11:2574. [PMID: 37761015 PMCID: PMC10526035 DOI: 10.3390/biomedicines11092574] [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: 08/01/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
There is a bidirectional relationship between the heart and the gut. The gut microbiota, the community of gut micro-organisms themselves, is an excellent gut-homeostasis keeper since it controls the growth of potentially harmful bacteria and protects the microbiota environment. There is evidence suggesting that a diet rich in fatty acids can be metabolized and converted by gut microbiota and hepatic enzymes to trimethyl-amine N-oxide (TMAO), a product that is associated with atherogenesis, platelet dysfunction, thrombotic events, coronary artery disease, stroke, heart failure (HF), and, ultimately, death. HF, by inducing gut ischemia, congestion, and, consequently, gut barrier dysfunction, promotes the intestinal leaking of micro-organisms and their products, facilitating their entrance into circulation and thus stimulating a low-grade inflammation associated with an immune response. Drugs used for HF may alter the gut microbiota, and, conversely, gut microbiota may modify the pharmacokinetic properties of the drugs. The modification of lifestyle based mainly on exercise and a Mediterranean diet, along with the use of pre- or probiotics, may be beneficial for the gut microbiota environment. The potential role of gut microbiota in HF development and progression is the subject of this review.
Collapse
Affiliation(s)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece
| | - Filippos Triposkiadis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| |
Collapse
|
9
|
Ren J, Li H, Zeng G, Pang B, Wang Q, Wei J. Gut microbiome-mediated mechanisms in aging-related diseases: are probiotics ready for prime time? Front Pharmacol 2023; 14:1178596. [PMID: 37324466 PMCID: PMC10267478 DOI: 10.3389/fphar.2023.1178596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
Chronic low-grade inflammation affects health and is associated with aging and age-related diseases. Dysregulation of the gut flora is an important trigger for chronic low-grade inflammation. Changes in the composition of the gut flora and exposure to related metabolites have an effect on the inflammatory system of the host. This results in the development of crosstalk between the gut barrier and immune system, contributing to chronic low-grade inflammation and impairment of health. Probiotics can increase the diversity of gut microbiota, protect the gut barrier, and regulate gut immunity, thereby reducing inflammation. Therefore, the use of probiotics is a promising strategy for the beneficial immunomodulation and protection of the gut barrier through gut microbiota. These processes might positively influence inflammatory diseases, which are common in the elderly.
Collapse
Affiliation(s)
- Jing Ren
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Huimin Li
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guixing Zeng
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Boxian Pang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Qiuhong Wang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Wei
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Sturov NV, Popov SV, Belikov II. Gut Microbiota and the Ways to Correct it in Chronic Kidney Disease. Indian J Nephrol 2023; 33:162-169. [PMID: 37448901 PMCID: PMC10337223 DOI: 10.4103/ijn.ijn_469_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 07/15/2023] Open
Abstract
Approximately 13% of the Russian population suffers from chronic kidney disease (CKD). Such a high prevalence of the disease, as well as the complexity and high cost of renal replacement therapy, explain the need for developing and implementing new approaches to treat patients at the pre-dialysis stages. The data collected in recent decades highlight the importance of gut microbiota in the progression of CKD. This review provides information about the microbiota composition in healthy individuals and patients with CKD and discusses the mechanisms of interaction in the intestine-kidney system. The article also presents the specifics of the violation of gut microbiota (GM) and correction thereof in CKD.
Collapse
Affiliation(s)
- Nikolay V. Sturov
- Peoples’ Friendship University of Russia (RUDN University), Department of General Practice, 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Sergey V. Popov
- Peoples’ Friendship University of Russia (RUDN University), Department of General Practice, 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Igor I. Belikov
- Peoples’ Friendship University of Russia (RUDN University), Department of General Practice, 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| |
Collapse
|
11
|
Wu X, Zhao L, Zhang Y, Li K, Yang J. The role and mechanism of the gut microbiota in the development and treatment of diabetic kidney disease. Front Physiol 2023; 14:1166685. [PMID: 37153213 PMCID: PMC10160444 DOI: 10.3389/fphys.2023.1166685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic kidney disease (DKD) is a common complication in patients with diabetes mellitus (DM). Increasing evidence suggested that the gut microbiota participates in the progression of DKD, which is involved in insulin resistance, renin-angiotensin system (RAS) activation, oxidative stress, inflammation and immunity. Gut microbiota-targeted therapies including dietary fiber, supplementation with probiotics or prebiotics, fecal microbiota transplantation and diabetic agents that modulate the gut microbiota, such as metformin, glucagon-like peptide-1 (GLP-1) receptor agonists, dipeptidyl peptidase-4 (DPP-4) inhibitors, and sodium-glucose transporter-2 (SGLT-2) inhibitors. In this review, we summarize the most important findings about the role of the gut microbiota in the pathogenesis of DKD and the application of gut microbiota-targeted therapies.
Collapse
Affiliation(s)
- Xiaofang Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Zhao
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yujiang Zhang
- Department of Nephrology, Chongqing Jiangjin Second People’s Hospital, Chongqing, China
| | - Kailong Li
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jurong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Jurong Yang,
| |
Collapse
|
12
|
Masenga SK, Hamooya B, Hangoma J, Hayumbu V, Ertuglu LA, Ishimwe J, Rahman S, Saleem M, Laffer CL, Elijovich F, Kirabo A. Recent advances in modulation of cardiovascular diseases by the gut microbiota. J Hum Hypertens 2022; 36:952-959. [PMID: 35469059 PMCID: PMC9649420 DOI: 10.1038/s41371-022-00698-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/29/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
Abstract
The gut microbiota has recently gained attention due to its association with cardiovascular health, cancers, gastrointestinal disorders, and non-communicable diseases. One critical question is how the composition of the microbiota contributes to cardiovascular diseases (CVDs). Insightful reviews on the gut microbiota, its metabolites and the mechanisms that underlie its contribution to CVD are limited. Hence, the aim of this review was to describe linkages between the composition of the microbiota and CVD, CVD risk factors such as hypertension, diet, ageing, and sex differences. We have also highlighted potential therapies for improving the composition of the gut microbiota, which may result in better cardiovascular health.
Collapse
Affiliation(s)
- Sepiso K Masenga
- Mulungushi University, School of Medicine and Health Sciences, HAND Research Group, Livingstone, Zambia
| | - Benson Hamooya
- Mulungushi University, School of Medicine and Health Sciences, HAND Research Group, Livingstone, Zambia
| | - Joy Hangoma
- Mulungushi University, School of Medicine and Health Sciences, HAND Research Group, Livingstone, Zambia
| | - Valerie Hayumbu
- Mulungushi University, School of Medicine and Health Sciences, HAND Research Group, Livingstone, Zambia
| | - Lale A Ertuglu
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - Jeanne Ishimwe
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - Sharla Rahman
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - Mohammad Saleem
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - Cheryl L Laffer
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - Fernando Elijovich
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA
| | - Annet Kirabo
- Vanderbilt University Medical Center, Department of Medicine, Nashville, TN, USA.
| |
Collapse
|
13
|
Andújar-Tenorio N, Prieto I, Cobo A, Martínez-Rodríguez AM, Hidalgo M, Segarra AB, Ramírez M, Gálvez A, Martínez-Cañamero M. High fat diets induce early changes in gut microbiota that may serve as markers of ulterior altered physiological and biochemical parameters related to metabolic syndrome. Effect of virgin olive oil in comparison to butter. PLoS One 2022; 17:e0271634. [PMID: 35972974 PMCID: PMC9380944 DOI: 10.1371/journal.pone.0271634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
Butter and virgin olive oil (EVOO) are two fats differing in their degree of saturation and insaponifiable fraction. EVOO, enriched in polyphenols and other minority components, exerts a distinct effect on health. Using next generation sequencing, we have studied early and long-term effects of both types of fats on the intestinal microbiota of mice, finding significant differences between the two diets in the percentage of certain bacterial taxa, correlating with hormonal, physiological and metabolic parameters in the host. These correlations are not only concomitant, but most noticeably some of the changes detected in the microbial percentages at six weeks are correlating with changes in physiological values detected later, at twelve weeks. Desulfovibrionaceae/Desulfovibrio/D. sulfuricans stand out by presenting at six weeks a statistically significant higher percentage in the butter-fed mice with respect to the EVOO group, correlating with systolic blood pressure, food intake, water intake and insulin at twelve weeks. This not only suggests an early implication in the probability of developing altered physiological and biochemical responses later on in the host lifespan, but also opens the possibility of using this genus as a marker in the risk of suffering different pathologies in the future.
Collapse
Affiliation(s)
- Natalia Andújar-Tenorio
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Isabel Prieto
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Antonio Cobo
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | | | - Marina Hidalgo
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Ana Belén Segarra
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Manuel Ramírez
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Antonio Gálvez
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | | |
Collapse
|
14
|
Liu T, Lu X, Gao W, Zhai Y, Li H, Li S, Yang L, Ma F, Zhan Y, Mao H. Cardioprotection effect of Yiqi-Huoxue-Jiangzhuo formula in a chronic kidney disease mouse model associated with gut microbiota modulation and NLRP3 inflammasome inhibition. Biomed Pharmacother 2022; 152:113159. [PMID: 35661533 DOI: 10.1016/j.biopha.2022.113159] [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: 03/25/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The pathogenesis and treatment of cardiovascular disease mediated by chronic kidney disease (CKD) are key research questions. Specifically, the mechanisms underlying the cardiorenal protective effect of Yiqi-Huoxue-Jiangzhuo formula (YHJF), a traditional Chinese herbal medicine, have not yet been clarified. METHODS A classical CKD mouse model was constructed by 5/6 nephrectomy (Nx) to study the effects of YHJF intervention on 5/6 Nx mice cardiorenal function, gut microbial composition, gut-derived metabolites, and NLRP3 inflammasome pathways. RESULTS YHJF improved cardiac dysfunction and reversed left ventricular hypertrophy, myocardial hypertrophy, and interstitial fibrosis in 5/6 Nx mice. In addition, YHJF inhibited activation of the NLRP3 inflammasome and downregulated the expression of TNF-α and IL-1β both in the heart and serum; reconstitution of the intestinal flora imbalance was also found in 5/6 Nx mice treated with YHJF. Spearman's correlation and redundancy analyses showed that changes in the intestinal flora of 5/6 Nx mice were related to clinical phenotype and serum inflammatory levels. CONCLUSIONS Treatment with YHJF effectively protected the heart function of 5/6 Nx mice; this effect was attributed to inhibition of NLRP3 inflammasome activation and regulation of intestinal microbial composition and derived metabolites. YHJF has potential for improving intestinal flora imbalance and gut-derived toxin accumulation in patients with CKD, thereby preventing cardiovascular complications.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaoguang Lu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wenya Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuanyuan Zhai
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing 100029, China
| | - Han Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shangheng Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
15
|
Chen R, Zhu D, Yang R, Wu Z, Xu N, Chen F, Zhang S, Chen H, Li M, Hou K. Gut microbiota diversity in middle-aged and elderly patients with end-stage diabetic kidney disease. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:750. [PMID: 35957707 PMCID: PMC9358493 DOI: 10.21037/atm-22-2926] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is the most common cause of end-stage renal disease (ESRD), but the mechanism between DKD and ESRD remains unclear. Some experts have put forward the "microbial-centered ESRD development theory", believing that the bacterial load caused by gut microecological imbalance and uremia toxin transfer are the core pathogenic links. The purpose of this study was to analyze the genomic characteristics of gut microbiota in patients with ESRD, specifically DKD or non-diabetic kidney disease (NDKD). METHODS In this cross-sectional study, patients with ESRD were recruited in a community, including 22 DKD patients and 22 NDKD patients matched using gender and age. Fecal samples of patients were collected for 16S rDNA sequencing and gut microbiota analysis. The distribution structure, diversity, and abundance of microflora in DKD patients were analyzed by constructing species evolutionary trees and analyzing alpha diversity, beta diversity, and linear discriminant analysis effect size (LEfSe). RESULTS The results of our study showed that there were statistically significant differences in the richness and species of gut microbiota at the total level between DKD patients and NDKD patients. The analysis of genus level between the two groups showed significant differences in 16 bacterial genera. Among them, Oscillibacter, Bilophila, UBA1819, Ruminococcaceae UCG-004, Anaerotruncus, Ruminococcaceae, and Ruminococcaceae NK4A214 bacteria in DKD patients were higher than those in NDKD patients. CONCLUSIONS 16S rDNA sequencing technology was used in this study to analyze the characteristics of intestinal flora in ESRD patients with or without diabetes. We found that there was a significant difference in the intestinal flora of ESRD patients caused by DKD and NDKD, suggesting that these may be potential causative bacteria for the development of ERSD in DKD patients.
Collapse
Affiliation(s)
- Rongping Chen
- School of Laboratory Medical and Biotechnology, Southern Medical University, Guangzhou, China
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Rui Yang
- Department of Endocrine and Metabolic Diseases, Southern Medical University, Guangzhou, China
| | - Zezhen Wu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Ningning Xu
- Department of Endocrine and Metabolic Diseases, Southern Medical University, Guangzhou, China
| | - Fengwu Chen
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Shuo Zhang
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Hong Chen
- Department of Endocrine and Metabolic Diseases, Southern Medical University, Guangzhou, China
| | - Ming Li
- School of Laboratory Medical and Biotechnology, Southern Medical University, Guangzhou, China
| | - Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Jinxia Community Health Service Centre, Shantou, China
| |
Collapse
|
16
|
Zhou F, Shao Q, Jia L, Cai C. Gut Microbiota Variations between Henoch-Schonlein Purpura and Henoch-Schonlein Purpura Nephritis. Gastroenterol Res Pract 2022; 2022:4003491. [PMID: 35462986 PMCID: PMC9023217 DOI: 10.1155/2022/4003491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background In China, little is known regarding the differences between children with Henoch-Schonlein purpura (HSP) and Henoch-Schonlein purpura nephritis (HSPN) concerning their gut microbiota. Methods We recruited 25 children with HSP, 25 children with HSPN, and 25 healthy children to investigate the differences. Fecal samples were collected and analyzed by sequencing the V3-V4 region of the 16S rRNA gene. The diversity of the fecal gut microbiota was compared between the patient groups. Results Rarefaction curves showed that the gut microbial diversity between the three groups differed significantly (P = 0.0224). The top five most abundant gut microbial genera were Bacteroides, Faecalibacterium, Prevotella, Ruminococcaceae, and Megamonas in children with HSP; Bacteroides, Faecalibacterium, Prevotella, Bifidobacterium, and Ruminococcaceae in children with HSPN; and Bacteroides, Prevotella, Faecalibacterium, Ruminococcaceae, and Bifidobacterium in healthy children. Children with HSP had the lowest Bifidobacterium abundance among the three groups (P < 0.05). Children with HSPN had a lower abundance of Akkermansia than children with HSP (P < 0.05), whereas children with HSPN had a higher Alistipes abundance than children with HSP (P < 0.05). Fecal microbial community composition did not differ significantly between groups (ANOSIM, R = -0.002, P = 0.46). Despite the small sample size, our results indicate that children with HSP or HSPN displayed dysbiosis of the gut microbiota. Conclusion This study provides valuable insights that will benefit the development of future microbe-based therapies to improve clinical outcomes or prevent the incidence of HSP or HSPN in children.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Rheumatoid Immune Nephrology, Hangzhou Children's Hospital, Hangzhou, 310014 Zhejiang, China
| | - Qimin Shao
- Department of Rheumatoid Immune Nephrology, Hangzhou Children's Hospital, Hangzhou, 310014 Zhejiang, China
| | - Lihong Jia
- Department of Rheumatoid Immune Nephrology, Hangzhou Children's Hospital, Hangzhou, 310014 Zhejiang, China
| | - Chunyan Cai
- Department of Rheumatoid Immune Nephrology, Hangzhou Children's Hospital, Hangzhou, 310014 Zhejiang, China
| |
Collapse
|
17
|
Xu X, Chen J, Lv H, Xi Y, Ying A, Hu X. Molecular mechanism of Pyrrosia lingua in the treatment of nephrolithiasis: Network pharmacology analysis and in vivo experimental verification. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 98:153929. [PMID: 35104754 DOI: 10.1016/j.phymed.2022.153929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/27/2021] [Accepted: 01/03/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Evidence exists reporting that Pyrrosia lingua (PL, Xinhui Pharmaceutical, Polypodiaceae) alleviates nephrolithiasis in rat models. The precipitation of calcium oxalate may result in kidney stones, and the intestinal microbiota is critical for oxalate metabolism. Therefore, we attempt to delineate the molecular mechanism underlying the effect of PL on nephrolithiasis and its association with gut microbiota. METHODS Following differential flora analysis in gutMEGA, the network relationship of PL and nephrolithiasis was analyzed based on the TCMSP, DisGeNET and STRING databases. Moreover, the kidney stone model rats were fed with different doses of PL powder and PL extract. In addition, metabolomics technology was employed to identify the active ingredients in PL extract and the microbial metabolites in rat feces. RESULTS The effect of PL on the nephrolithiasis was based on quercetin and kaempferol by mediating the toll-like receptor signaling pathway and regulating the expression levels of interleukin 6, tumor necrosis factor, mitogen activated protein kinase 8, and secreted phosphoprotein 1. PL significantly reduced the levels of urine oxalic acid, urine calcium, and osteopontin (OPN) levels in rat models of nephrolithiasis. Notably, PL extract decreased these two indicators to lower levels. Furthermore, contents of Oxalobacter formigenes, Bacteriodetes, Bifidobacterium and Fecalibacterium were obviously reduced after treatment with PL extract. CONCLUSION PL powder and its active extracts reduce the oxalate level in urine by regulating oxalate metabolism, thus ameliorating the damage of kidney tissues and preventing kidney stone formation. This study suggests the use of PL and its extracts as an alternative source of promising agents that might directly or indirectly inhibit the progression of kidney stone diseases.
Collapse
Affiliation(s)
- Xiangwei Xu
- Department of Pharmacy, Yongkang First People's Hospital Affiliated to Hangzhou Medical College, Yongkang 321300, PR China
| | - Jun Chen
- Department of Pharmacy, Yongkang First People's Hospital Affiliated to Hangzhou Medical College, Yongkang 321300, PR China
| | - Haiou Lv
- Department of Urology Surgery, Yongkang First people's Hospital Affiliated to Hangzhou Medical College, 599 jinshan West Road, Dongcheng Street, Yongkang, Zhejiang Province 321300, PR China
| | - Yiyuan Xi
- School of Pharmacy, Wenzhou Medical University, PR China
| | - Aiying Ying
- Department of Urology Surgery, Yongkang First people's Hospital Affiliated to Hangzhou Medical College, 599 jinshan West Road, Dongcheng Street, Yongkang, Zhejiang Province 321300, PR China
| | - Xiang Hu
- Department of Urology Surgery, Yongkang First people's Hospital Affiliated to Hangzhou Medical College, 599 jinshan West Road, Dongcheng Street, Yongkang, Zhejiang Province 321300, PR China.
| |
Collapse
|
18
|
Maiuolo J, Carresi C, Gliozzi M, Mollace R, Scarano F, Scicchitano M, Macrì R, Nucera S, Bosco F, Oppedisano F, Ruga S, Coppoletta AR, Guarnieri L, Cardamone A, Bava I, Musolino V, Paone S, Palma E, Mollace V. The Contribution of Gut Microbiota and Endothelial Dysfunction in the Development of Arterial Hypertension in Animal Models and in Humans. Int J Mol Sci 2022; 23:ijms23073698. [PMID: 35409057 PMCID: PMC8999124 DOI: 10.3390/ijms23073698] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
The maintenance of the physiological values of blood pressure is closely related to unchangeable factors (genetic predisposition or pathological alterations) but also to modifiable factors (dietary fat and salt, sedentary lifestyle, overweight, inappropriate combinations of drugs, alcohol abuse, smoking and use of psychogenic substances). Hypertension is usually characterized by the presence of a chronic increase in systemic blood pressure above the threshold value and is an important risk factor for cardiovascular disease, including myocardial infarction, stroke, micro- and macro-vascular diseases. Hypertension is closely related to functional changes in the endothelium, such as an altered production of vasoconstrictive and vasodilator substances, which lead to an increase in vascular resistance. These alterations make the endothelial tissue unresponsive to autocrine and paracrine stimuli, initially determining an adaptive response, which over time lead to an increase in risk or disease. The gut microbiota is composed of a highly diverse bacterial population of approximately 1014 bacteria. A balanced intestinal microbiota preserves the digestive and absorbent functions of the intestine, protecting from pathogens and toxic metabolites in the circulation and reducing the onset of various diseases. The gut microbiota has been shown to produce unique metabolites potentially important in the generation of hypertension and endothelial dysfunction. This review highlights the close connection between hypertension, endothelial dysfunction and gut microbiota.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Laboratory of Pharmaceutical Biology, in IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy;
- Correspondence: (J.M.); (M.G.)
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Correspondence: (J.M.); (M.G.)
| | - Rocco Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Roberta Macrì
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Irene Bava
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Vincenzo Musolino
- Laboratory of Pharmaceutical Biology, in IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy;
| | - Sara Paone
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Ernesto Palma
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
| |
Collapse
|
19
|
Nagase N, Ikeda Y, Tsuji A, Kitagishi Y, Matsuda S. Efficacy of probiotics on the modulation of gut microbiota in the treatment of diabetic nephropathy. World J Diabetes 2022; 13:150-160. [PMID: 35432750 PMCID: PMC8984564 DOI: 10.4239/wjd.v13.i3.150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/21/2021] [Accepted: 02/13/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease, and therapeutic options for preventing its progression are insufficient. The number of patients with DN has been increasing in Asian countries because of westernization of dietary lifestyle, which may be associated with the following changes in gut microbiota. Alterations in the gut microbiota composition can lead to an imbalanced gastrointestinal environment that promotes abnormal production of metabolites and/or inflammatory status. Functional microenvironments of the gut could be changed in the different stages of DN. In particular, altered levels of short chain fatty acids, D-amino acids, and reactive oxygen species biosynthesis in the gut have been shown to be relevant to the pathogenesis of the DN. So far, evidence suggests that the gut microbiota may play a key role in determining networks in the development of DN. Interventions directing the gut microbiota deserve further investigation as a new protective therapy in DN. In this review, we discuss the potential roles of the gut microbiota and future perspectives in the protection and/or treatment of kidneys.
Collapse
Affiliation(s)
- Nozomi Nagase
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Yuka Ikeda
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Ai Tsuji
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| |
Collapse
|
20
|
Intestinal Microbiota as a Contributor to Chronic Inflammation and Its Potential Modifications. Nutrients 2021; 13:nu13113839. [PMID: 34836095 PMCID: PMC8618457 DOI: 10.3390/nu13113839] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is a crucial factor in maintaining homeostasis. The presence of commensal microorganisms leads to the stimulation of the immune system and its maturation. In turn, dysbiosis with an impaired intestinal barrier leads to accelerated contact of microbiota with the host’s immune cells. Microbial structural parts, i.e., pathogen-associated molecular patterns (PAMPs), such as flagellin (FLG), peptidoglycan (PGN), lipoteichoic acid (LTA), and lipopolysaccharide (LPS), induce inflammation via activation of pattern recognition receptors. Microbial metabolites can also develop chronic low-grade inflammation, which is the cause of many metabolic diseases. This article aims to systematize information on the influence of microbiota on chronic inflammation and the benefits of microbiota modification through dietary changes, prebiotics, and probiotic intake. Scientific research indicates that the modification of the microbiota in various disease states can reduce inflammation and improve the metabolic profile. However, since there is no pattern for a healthy microbiota, there is no optimal way to modify it. The methods of influencing microbiota should be adapted to the type of dysbiosis. Although there are studies on the microbiota and its effects on inflammation, this subject is still relatively unknown, and more research is needed in this area.
Collapse
|
21
|
Vasileva VY, Sultanova RF, Sudarikova AV, Ilatovskaya DV. Insights Into the Molecular Mechanisms of Polycystic Kidney Diseases. Front Physiol 2021; 12:693130. [PMID: 34566674 PMCID: PMC8456103 DOI: 10.3389/fphys.2021.693130] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant (AD) and autosomal recessive (AR) polycystic kidney diseases (PKD) are severe multisystem genetic disorders characterized with formation and uncontrolled growth of fluid-filled cysts in the kidney, the spread of which eventually leads to the loss of renal function. Currently, there are no treatments for ARPKD, and tolvaptan is the only FDA-approved drug that alleviates the symptoms of ADPKD. However, tolvaptan has only a modest effect on disease progression, and its long-term use is associated with many side effects. Therefore, there is still a pressing need to better understand the fundamental mechanisms behind PKD development. This review highlights current knowledge about the fundamental aspects of PKD development (with a focus on ADPKD) including the PC1/PC2 pathways and cilia-associated mechanisms, major molecular cascades related to metabolism, mitochondrial bioenergetics, and systemic responses (hormonal status, levels of growth factors, immune system, and microbiome) that affect its progression. In addition, we discuss new information regarding non-pharmacological therapies, such as dietary restrictions, which can potentially alleviate PKD.
Collapse
Affiliation(s)
| | - Regina F Sultanova
- Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia.,Department of Physiology, Augusta University, Augusta, GA, United States
| | | | | |
Collapse
|
22
|
Zhao X, Zhong X, Liu X, Wang X, Gao X. Therapeutic and Improving Function of Lactobacilli in the Prevention and Treatment of Cardiovascular-Related Diseases: A Novel Perspective From Gut Microbiota. Front Nutr 2021; 8:693412. [PMID: 34164427 PMCID: PMC8215129 DOI: 10.3389/fnut.2021.693412] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
The occurrence and development of cardiovascular-related diseases are associated with structural and functional changes in gut microbiota (GM). The accumulation of beneficial gut commensals contributes to the improvement of cardiovascular-related diseases. The cardiovascular-related diseases that can be relieved by Lactobacillus supplementation, including hypercholesterolemia, atherosclerosis, myocardial infarction, heart failure, type 2 diabetes mellitus, and obesity, have expanded. As probiotics, lactobacilli occupy a substantial part of the GM and play important functional roles through various GM-derived metabolites. Lactobacilli ultimately have a beneficial impact on lipid metabolism, inflammatory factors, and oxidative stress to relieve the symptoms of cardiovascular-related diseases. However, the axis and cellular process of gut commensal Lactobacillus in improving cardiovascular-related diseases have not been fully elucidated. Additionally, Lactobacillus strains produce diverse antimicrobial peptides, which help maintain intestinal homeostasis and ameliorate cardiovascular-related diseases. These strains are a field that needs to be further investigated immediately. Thus, this review demonstrated the mechanisms and summarized the evidence of the benefit of Lactobacillus strain supplementation from animal studies and human clinical trials. We also highlighted a broad range of lactobacilli candidates with therapeutic capability by mining their metabolites. Our study provides instruction in the development of lactobacilli as a functional food to improve cardiovascular-related diseases.
Collapse
Affiliation(s)
- Xin Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinqin Zhong
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoying Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
23
|
Lin W, Jiang C, Yu H, Wang L, Li J, Liu X, Wang L, Yang H. The effects of Fushen Granule on the composition and function of the gut microbiota during Peritoneal Dialysis-Related Peritonitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 86:153561. [PMID: 33857850 DOI: 10.1016/j.phymed.2021.153561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Peritoneal dialysis (PD) is an acknowledged treatment for patients with irreversible kidney failure. The treatment usually causes peritoneal dialysis-related peritonitis (PDRP), a common complication of PD that can lead to inadequate dialysis, gastrointestinal dysfunction, and even death. Recent studies indicated that Fushen Granule (FSG), a Chinese herbal formula, improves the treatment of PD. However, the mechanism of how FSG plays its role in the improvement is still unclear. Gut microbiota has been closely related to the development of various diseases. We carried out a randomized controlled trial to assess whether FSG can modulate the gut microbiota during PDRP treatment. METHODS Forty-two PDRP patients were recruited into the clinical trial, and they were randomly divided into control(CON), probiotics(PRO) or Fushen granule group(FSG). To check whether FSG improve the PD treatment, we assessed the clinical parameters, including albumin(ALB), hemoglobin(HGB), blood urea nitrogen(BUN) and creatinine(CR). Fecal samples were collected before hospitalization and discharge, and stored at -80°C within 1 hour. And we assessed the microbial population and function by applying the 16S rRNA gene sequencing and functional enrichment analysis. RESULTS Compared to control group, ALB is improved in both probiotics and FSG groups, while HGB is increased but BUN and CR is reduced in FSG group. Sequencing of 16S rRNA genes revealed that FSG and PRO affected the composition of the microbial community. FSG significantly increased a abundant represented by Bacteroides, Megamonas and Rothia, which was significantly correlated with the improvements in carbohydrate and amino acid metabolism. CONCLUSIONS This study demonstrates that FSG ameliorates the nutritional status and improves the quality of life by enriching beneficial bacteria associated with metabolism. These results indicate that FSG as alternative medicine is a promising treatment for patients with PDRP.
Collapse
Affiliation(s)
- Wei Lin
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, PR China
| | - Chen Jiang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, PR China
| | - Hangxing Yu
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, PR China
| | - Lingling Wang
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jiaqi Li
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, PR China
| | - Xinyue Liu
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, PR China
| | - Lingyun Wang
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hongtao Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, PR China.
| |
Collapse
|
24
|
Taguchi K, Fukami K, Elias BC, Brooks CR. Dysbiosis-Related Advanced Glycation Endproducts and Trimethylamine N-Oxide in Chronic Kidney Disease. Toxins (Basel) 2021; 13:361. [PMID: 34069405 PMCID: PMC8158751 DOI: 10.3390/toxins13050361] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a public health concern that affects approximately 10% of the global population. CKD is associated with poor outcomes due to high frequencies of comorbidities such as heart failure and cardiovascular disease. Uremic toxins are compounds that are usually filtered and excreted by the kidneys. With the decline of renal function, uremic toxins are accumulated in the systemic circulation and tissues, which hastens the progression of CKD and concomitant comorbidities. Gut microbial dysbiosis, defined as an imbalance of the gut microbial community, is one of the comorbidities of CKD. Meanwhile, gut dysbiosis plays a pathological role in accelerating CKD progression through the production of further uremic toxins in the gastrointestinal tracts. Therefore, the gut-kidney axis has been attracting attention in recent years as a potential therapeutic target for stopping CKD. Trimethylamine N-oxide (TMAO) generated by gut microbiota is linked to the progression of cardiovascular disease and CKD. Also, advanced glycation endproducts (AGEs) not only promote CKD but also cause gut dysbiosis with disruption of the intestinal barrier. This review summarizes the underlying mechanism for how gut microbial dysbiosis promotes kidney injury and highlights the wide-ranging interventions to counter dysbiosis for CKD patients from the view of uremic toxins such as TMAO and AGEs.
Collapse
Affiliation(s)
- Kensei Taguchi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (B.C.E.); (C.R.B.)
| | - Kei Fukami
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan;
| | - Bertha C. Elias
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (B.C.E.); (C.R.B.)
| | - Craig R. Brooks
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (B.C.E.); (C.R.B.)
| |
Collapse
|
25
|
Quintela BCSF, Carioca AAF, de Oliveira JGR, Fraser SDS, da Silva Junior GB. Dietary patterns and chronic kidney disease outcomes: A systematic review. Nephrology (Carlton) 2021; 26:603-612. [PMID: 33864650 DOI: 10.1111/nep.13883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 07/09/2020] [Accepted: 04/14/2021] [Indexed: 01/11/2023]
Abstract
Chronic kidney disease (CKD) is a serious public health problem and its prevalence is growing in many countries, often related to issues resulting from the lifestyle in growing economies and the population's life expectancy. Nutritional therapy is a beneficial but still neglected strategy for preventing CKD and delaying disease progression. The aim of this study was to assess the association of dietary patterns with CKD development and progression. Observational studies conducted in adult humans and the correlation between the adopted dietary pattern and prevalent and incident cases of CKD were assessed. A significant association was observed between unhealthy dietary patterns and an increased risk of developing or worsening CKD, as well as an adverse effect. Whereas healthy eating patterns characterized by the consumption of fruit, vegetables and dietary fibre showed nephroprotective outcomes.
Collapse
Affiliation(s)
| | | | | | - Simon D S Fraser
- Academic Unit of Primary Care and Population Science, Faculty of Medicine, University of Southampton, Southampton, Hampshire, United Kingdom
| | | |
Collapse
|
26
|
Banaszewska B, Siakowska M, Chudzicka-Strugala I, Chang RJ, Pawelczyk L, Zwozdziak B, Spaczynski R, Duleba AJ. Elevation of markers of endotoxemia in women with polycystic ovary syndrome. Hum Reprod 2021; 35:2303-2311. [PMID: 32869098 DOI: 10.1093/humrep/deaa194] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Is polycystic ovary syndrome (PCOS) associated with an elevation of markers of endotoxemia? SUMMARY ANSWER In women with PCOS serum levels of lipopolysaccharides (LPS), the LPS to high-density lipoprotein (HDL) ratio and LPS-binding protein (LBP) are significantly greater than those of normal control subjects. WHAT IS KNOWN ALREADY Mononuclear cells from women with PCOS respond excessively to LPS by releasing pro-inflammatory cytokines. In rat ovarian theca-interstitial cell cultures LPS stimulates androgen production. STUDY DESIGN, SIZE, DURATION Cross-sectional study comparing markers of endotoxemia in women with PCOS (n = 62), healthy ovulatory women with polycystic ovary morphology (PCOM, n = 39) and a control group of healthy ovulatory women without PCOM [normal (NL), n = 43]. PARTICIPANTS/MATERIALS, SETTING, METHODS LPS was measured using a chromogenic assay. LBP was measured by ELISA. Total cholesterol and lipids were measured using a homogeneous enzyme colorimetric method. Androgens, gonadotrophins, prolactin, insulin, high-sensitivity C-reactive protein (hs-CRP) and sex hormone-binding globulin were determined by electrochemiluminescence assays. Glucose was measured using an enzymatic reference method with hexokinase. MAIN RESULTS AND THE ROLE OF CHANCE Women with PCOS, when compared with NL subjects, had a significantly higher mean LPS (P = 0.045), LPS/HDL ratio (P = 0.007) and LBP (P = 0.01). Women with PCOM had intermediate levels of markers of endotoxemia. Comparison among all groups revealed that markers of endotoxemia correlated positively with testosterone level, ovarian volume, number of antral follicles and hirsutism score, but negatively with the number of spontaneous menses per year. In multiple regression analysis, all measures of endotoxemia correlated independently and positively with hs-CRP and with ovarian volume. LIMITATIONS, REASONS FOR CAUTION This cross-sectional study reveals that markers of endotoxemia are associated with several clinical features observed in women with PCOS. However, responsible mechanisms and causation remain unknown. Steroid quantification was carried out by electrochemiluminescence assays and not by the current gold standard: liquid chromatography-mass spectrometry. Hence, the relationship of endotoxemia with features of PCOS and the extent to which endotoxemia contributes to reproductive and metabolic dysfunction warrants further investigation. WIDER IMPLICATIONS OF THE FINDINGS This study reveals the novel observation that markers of endotoxemia are elevated in young and otherwise healthy women with PCOS without significant metabolic dysfunction. Moreover, the association of clinical and endocrine markers of PCOS with those of endotoxemia may represent a pathophysiologic link to reproductive dysfunction as well as metabolic and long-term cardiovascular risks associated with this disorder. STUDY FUNDING/COMPETING INTEREST(S) Intramural funding from Poznan University of Medical Sciences. The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Beata Banaszewska
- Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Martyna Siakowska
- Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - R Jeffrey Chang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Leszek Pawelczyk
- Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Barbara Zwozdziak
- Department of Medical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Robert Spaczynski
- Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Antoni J Duleba
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
Rysz J, Franczyk B, Ławiński J, Olszewski R, Ciałkowska-Rysz A, Gluba-Brzózka A. The Impact of CKD on Uremic Toxins and Gut Microbiota. Toxins (Basel) 2021; 13:toxins13040252. [PMID: 33807343 PMCID: PMC8067083 DOI: 10.3390/toxins13040252] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 12/11/2022] Open
Abstract
Numerous studies have indicated that the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD) is strictly associated with the accumulation of toxic metabolites in blood and other metabolic compartments. This accumulation was suggested to be related to enhanced generation of toxins from the dysbiotic microbiome accompanied by their reduced elimination by impaired kidneys. Intestinal microbiota play a key role in the accumulation of uremic toxins due to the fact that numerous uremic solutes are generated in the process of protein fermentation by colonic microbiota. Some disease states, including CKD, are associated with the presence of dysbiosis, which can be defined as an "imbalanced intestinal microbial community with quantitative and qualitative changes in the composition and metabolic activities of the gut microbiota". The results of studies have confirmed the altered composition and functions of gut microbial community in chronic kidney disease. In the course of CKD protein-bound uremic toxins, including indoxyl sulfate, p-cresyl glucuronide, p-cresyl sulfate and indole-3-acetic acid are progressively accumulated. The presence of chronic kidney disease may be accompanied by the development of intestinal inflammation and epithelial barrier impairment leading to hastened systemic translocation of bacterial-derived uremic toxins and consequent oxidative stress injury to the kidney, cardiovascular and endocrine systems. These findings offer new therapeutic possibilities for the management of uremia, inflammation and kidney disease progression and the prevention of adverse outcomes in CKD patients. It seems that dietary interventions comprising prebiotics, probiotics, and synbiotics could pose a promising strategy in the management of uremic toxins in CKD.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Janusz Ławiński
- Department of Urology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszow, Poland;
| | - Robert Olszewski
- Department of Gerontology, Public Health and Didactics, Rheumatology and Rehabilitation, National Institute of Geriatrics, 02-637 Warsaw, Poland;
- Department of Ultrasound, Institute of Fundamental Technological Research, Polish Academy of Sciences, 02-637 Warsaw, Poland
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
- Correspondence: ; Tel.: +48-42-6393750
| |
Collapse
|
28
|
Thrombolome and Its Emerging Role in Chronic Kidney Diseases. Toxins (Basel) 2021; 13:toxins13030223. [PMID: 33803899 PMCID: PMC8003125 DOI: 10.3390/toxins13030223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of thromboembolic complications, including myocardial infarction, stroke, deep vein thrombosis, and pulmonary embolism. These complications lead to increased mortality. Evidence points to the key role of CKD-associated dysbiosis and its effect via the generation of gut microbial metabolites in inducing the prothrombotic phenotype. This phenomenon is known as thrombolome, a panel of intestinal bacteria-derived uremic toxins that enhance thrombosis via increased tissue factor expression, platelet hyperactivity, microparticles release, and endothelial dysfunction. This review discusses the role of uremic toxins derived from gut-microbiota metabolism of dietary tryptophan (indoxyl sulfate (IS), indole-3-acetic acid (IAA), kynurenine (KYN)), phenylalanine/tyrosine (p-cresol sulfate (PCS), p-cresol glucuronide (PCG), phenylacetylglutamine (PAGln)) and choline/phosphatidylcholine (trimethylamine N-oxide (TMAO)) in spontaneously induced thrombosis. The increase in the generation of gut microbial uremic toxins, the activation of aryl hydrocarbon (AhRs) and platelet adrenergic (ARs) receptors, and the nuclear factor kappa B (NF-κB) signaling pathway can serve as potential targets during the prevention of thromboembolic events. They can also help create a new therapeutic approach in the CKD population.
Collapse
|
29
|
Coutinho-Wolino KS, de F Cardozo LFM, de Oliveira Leal V, Mafra D, Stockler-Pinto MB. Can diet modulate trimethylamine N-oxide (TMAO) production? What do we know so far? Eur J Nutr 2021; 60:3567-3584. [PMID: 33533968 DOI: 10.1007/s00394-021-02491-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/08/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is a metabolite that has attracted attention due to its positive association with several chronic non-communicable diseases such as insulin resistance, atherosclerotic plaque formation, diabetes, cancer, heart failure, hypertension, chronic kidney disease, liver steatosis, cardiac fibrosis, endothelial injury, neural degeneration and Alzheimer's disease. TMAO production results from the fermentation by the gut microbiota of dietary nutrients such as choline and carnitine, which are transformed to trimethylamine (TMA) and converted into TMAO in the liver by flavin-containing monooxygenase 1 and 3 (FMO1 and FMO3). Considering that TMAO is involved in the development of many chronic diseases, strategies have been found to enhance a healthy gut microbiota. In this context, some studies have shown that nutrients and bioactive compounds from food can modulate the gut microbiota and possibly reduce TMAO production. OBJECTIVE This review has as main objective to discuss the studies that demonstrated the effects of food on the reduction of this harmful metabolite. METHODS All relevant articles until November 2020 were included. The articles were searched in Medline through PubMed. RESULTS Both the food is eaten acutely and chronically, by altering the nature of the gut microbiota, influencing colonic TMA production. Furthermore, hepatic production of TMAO by the flavin monooxygenases in the liver may also be influenced by phenolic compounds present in foods. CONCLUSION The evidence presented in this review shows that TMAO levels can be reduced by some bioactive compounds. However, it is crucial to notice that there is significant variation among the studies. Further clinical studies should be conducted to evaluate these dietary components' effectiveness, dose, and intervention time on TMAO levels and its precursors.
Collapse
Affiliation(s)
| | - Ludmila F M de F Cardozo
- Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Viviane de Oliveira Leal
- Division of Nutrition, Pedro Ernesto University Hospital, State University of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Denise Mafra
- Postgraduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil.,Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Medical Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Milena Barcza Stockler-Pinto
- Postgraduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil.,Postgraduate Program in Cardiovascular Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
30
|
Li DY, Wang Z, Jia X, Yan D, Shih DM, Hazen SL, Lusis AJ, Tang WHW. Loop Diuretics Inhibit Renal Excretion of Trimethylamine N-Oxide. JACC Basic Transl Sci 2021; 6:103-115. [PMID: 33665512 PMCID: PMC7907536 DOI: 10.1016/j.jacbts.2020.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 12/26/2022]
Abstract
Uremic retention solutes predominantly eliminate through the kidneys largely via specific efflux channels in the proximal renal tubules. For the first time, we demonstrated in vivo that renal tubular excretion of TMAO can be inhibited by concomitant loop diuretic administration via competition at the level of renal transporters. We further observed accumulation of TMAO in the renal parenchyma, which implied differential distributions of TMAO across various tissues and/or systems as a consequence of efflux channel control. Poorer outcomes in patients who receive long-term loop diuretic agents may therefore be associated with metabolic perturbations, such as retention of metabolites like TMAO, beyond impaired glomerular filtration.
This study demonstrates, for the first time, that renal tubular excretion of trimethylamine N-oxide (TMAO) is inhibited by concomitant loop diuretic administration. The observed marked accumulation in the renal parenchyma, and to lesser extent, plasma, implies differential distributions of TMAO across various tissues and/or systems as a consequence of efflux channel control. A better understanding of TMAO renal clearance and its potential interactions with current and future therapies in patients with heart failure are warranted.
Collapse
Affiliation(s)
- Daniel Y Li
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Medicine, Stanford University Medical Center, Stanford, California, USA
| | - Zeneng Wang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xun Jia
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Di Yan
- Department of Dermatology, New York University Langone Health, New York, New York, USA
| | - Diana M Shih
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California, USA
| | - Stanley L Hazen
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California, USA
| | - W H Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Clinical Genomics, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
31
|
Yang P, Li X, Yang W, He L, Yang L, Zhang X. A simple liquid chromatography/differential ion mobility spectrometry tandem mass spectrometry method for the determination of trimethylamine-N-oxide in human serum: An application in dialysis patients. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2021; 35:e8965. [PMID: 33002250 DOI: 10.1002/rcm.8965] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Trimethylamine-N-oxide (TMAO) is a potential indicator of cardiovascular disease and chronic kidney disorders. It is important to monitor the TMAO level in plasma or serum of hemodialysis patients. A simple liquid chromatography/differential ion mobility spectrometry tandem mass spectrometry (HPLC/DMS-MS/MS) method was established and validated for the determination of TMAO in the serum of hemodialysis patients. METHODS Chromatographic separation was performed on a Waters Atlantis HILIC silica column (2.1 × 50 mm, 3 μm). The gradient mobile phase consisted of 10 mM ammonium formate buffer and acetonitrile with 0.1% formic acid in both solvents. The serum sample was precipitated with acidic acetonitrile prior to HPLC/DMS-MS/MS analysis and TMAO-d9 was used as the internal standard. Data acquisition was performed in positive ion mode with a DMS system before the electrospray ionization source. The selected reaction monitoring transitions were m/z 76.0 → 58.0 and m/z 85.2 → 66.1 for TMAO and the internal standard, respectively. RESULTS Excellent linearity was observed over the calibration range 0.05-20 μg/mL (r2 > 0.995). The method was validated for good specificity and sensitivity. The inter-run and intra-run precision and accuracy were less than 13.6% and 10.7%, respectively. CONCLUSIONS We established a novel and robust HPLC/DMS-MS/MS method for the quantification of TMAO in human serum samples. The validated assay was simple, rapid, sensitive and reliable. The developed method could be applied to the assay of serum samples from patients with kidney disease who are undergoing hemodialysis.
Collapse
Affiliation(s)
- Ping Yang
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaona Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
| | - Wenling Yang
- Department of Nephrology, Peking University Third Hospital, Beijing, 100191, China
| | - Lian He
- Department of Nephrology, Peking University Third Hospital, Beijing, 100191, China
| | - Li Yang
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
| | - Xianhua Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
32
|
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: 3] [Impact Index Per Article: 0.6] [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
|
33
|
Bryniarski MA, Hoffman SEM, Yacoub R. The Gastrointestinal Microbiome in Chronic Renal Diseases. CURRENT ORAL HEALTH REPORTS 2020; 7:45-53. [DOI: 10.1007/s40496-020-00258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
34
|
Abstract
Renal injury, especially chronic kidney disease (CKD), is closely associated with gut microbiota. It is well known that renal injury development could cause enteric microbial compositional disruption. On the other hand, gut microbial composition, as well as their function, would directly influence the renal disease progression. Here, in the present chapter, we will summarize the crosstalk between intestinal microbiota and renal disease and discuss some potential therapeutic approaches based on this topic.
Collapse
|
35
|
Sabit H, Cevik E, Tombuloglu H. Colorectal cancer: The epigenetic role of microbiome. World J Clin Cases 2019; 7:3683-3697. [PMID: 31799293 PMCID: PMC6887622 DOI: 10.12998/wjcc.v7.i22.3683] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 10/23/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in men (746000 cases per year) and the second most common cancer in women globally (614000 cases per year). The incidence rate of CRC in developed countries (737000 cases per year) is higher than that in less developed countries (624000 cases per year). CRC can arise from genetic causes such as chromosomal instability and microsatellite instability. Several etiologic factors underlie CRC including age, diet, and lifestyle. Gut microbiota represent a proven cause of the disease, where they play pivotal roles in modulating and reshaping the host epigenome. Several active microbial metabolites have been found to drive carcinogenesis, invasion, and metastasis via modifying both the methylation landscape along with histone structure in intestinal cells. Gut microbiota, in response to diet, can exert both beneficial and harmful functions in humans, according to the intestinal balance of number and types of these bacteria. Although the intestinal microbial community is diverse among individuals, these microbes cumulatively produce 100-fold more proteins than the human genome itself, which calls for further studies to elaborate on the complicated interaction between these microorganisms and intestinal cells. Therefore, understanding the exact role that gut microbiota play in inducing CRC will help attain reliable strategies to precisely diagnose and treat this fatal disease.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Emre Cevik
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Huseyin Tombuloglu
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| |
Collapse
|
36
|
Zhao H, Chen L, Yang T, Feng YL, Vaziri ND, Liu BL, Liu QQ, Guo Y, Zhao YY. Aryl hydrocarbon receptor activation mediates kidney disease and renal cell carcinoma. J Transl Med 2019; 17:302. [PMID: 31488157 PMCID: PMC6727512 DOI: 10.1186/s12967-019-2054-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a well-known ligand-activated cytoplasmic transcription factor that contributes to cellular responses against environmental toxins and carcinogens. AhR is activated by a range of structurally diverse compounds from the environment, microbiome, natural products, and host metabolism, suggesting that AhR possesses a rather promiscuous ligand binding site. Increasing studies have indicated that AhR can be activated by a variety of endogenous ligands and induce the expression of a battery of genes. AhR regulates a variety of physiopathological events, including cell proliferation, differentiation, apoptosis, adhesion and migration. These new roles have expanded our understanding of the AhR signalling pathways and endogenous metabolites interacting with AhR under homeostatic and pathological conditions. Recent studies have demonstrated that AhR is linked to cardiovascular disease (CVD), chronic kidney disease (CKD) and renal cell carcinoma (RCC). In this review, we summarize gut microbiota-derived ligands inducing AhR activity in patients with CKD, CVD, diabetic nephropathy and RCC that may provide a new diagnostic and prognostic approach for complex renal damage. We further highlight polyphenols from natural products as AhR agonists or antagonists that regulate AhR activity. A better understanding of structurally diverse polyphenols and AhR biological activities would allow us to illuminate their molecular mechanism and discover potential therapeutic strategies targeting AhR activation.
Collapse
Affiliation(s)
- Hui Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Lin Chen
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Tian Yang
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Ya-Long Feng
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, CA, 92897, USA
| | - Bao-Li Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Qing-Quan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Yan Guo
- Department of Internal Medicine, University of New Mexico, Albuquerque, 87131, USA
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China.
| |
Collapse
|
37
|
Kwant CT, Ruiter G, Vonk Noordegraaf A. Malnutrition in pulmonary arterial hypertension: a possible role for dietary intervention. Curr Opin Pulm Med 2019; 25:405-409. [PMID: 31365373 PMCID: PMC6693930 DOI: 10.1097/mcp.0000000000000608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The last decade's progress has been made in the pharmacological treatment of pulmonary arterial hypertension (PAH). The role of nutrition in relation to quality of life in this group of patients is not investigated yet. In addition to avoiding salt and high-fluid intake based on left heart failure diet, there is no evidence-based diet recommendation for PAH. RECENT FINDINGS It was recently demonstrated that patients with PAH suffer from malnutrition resulting in iron and vitamin D deficiency and glucose/insulin resistance. Recent experimental studies suggest that besides reduced malabsorption of important nutrients, the microbiome of the gut is also less diverse in PAH. In this review, we summarize the current knowledge on malnutrition and dietary intake in PAH. We discuss the possible underlying mechanisms and discuss novel therapeutic interventions validated in patients with left heart failure. SUMMARY Large-scaled studies on dietary interventions are needed in PAH.
Collapse
Affiliation(s)
- Chermaine T Kwant
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
38
|
Perna AF, Glorieux G, Zacchia M, Trepiccione F, Capolongo G, Vigorito C, Anishchenko E, Ingrosso D. The role of the intestinal microbiota in uremic solute accumulation: a focus on sulfur compounds. J Nephrol 2019; 32:733-740. [PMID: 30673975 DOI: 10.1007/s40620-019-00589-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022]
Abstract
The gut microbiota is considered to be a novel important factor to take into account in the pathogenesis of chronic kidney disease and uremia. Much attention has been paid to specific uremic retention solutes of microbial origin, such as indoxyl sulfate, p-cresyl sulfate, and trimethylamine-N-oxide. However, other novel less well studied compounds, such as hydrogen sulfide and related sulfur metabolites (sulfane sulfur, lanthionine, etc.), should be included in a more comprehensive appraisal of this topic, in light of the potential therapeutic opportunities for the future.
Collapse
Affiliation(s)
- Alessandra F Perna
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5, Bldg 17, 80131, Naples, Italy.
| | - Griet Glorieux
- Nephrology Section, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Miriam Zacchia
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5, Bldg 17, 80131, Naples, Italy
| | - Francesco Trepiccione
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5, Bldg 17, 80131, Naples, Italy
| | - Giovanna Capolongo
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5, Bldg 17, 80131, Naples, Italy
| | - Carmela Vigorito
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5, Bldg 17, 80131, Naples, Italy
| | - Evgeniya Anishchenko
- First Division of Nephrology, Department of Translational Medical Sciences, School of Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5, Bldg 17, 80131, Naples, Italy
| | - Diego Ingrosso
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Via Luigi de Crecchio 7, 80138, Naples, Italy
| |
Collapse
|
39
|
Chen YY, Chen DQ, Chen L, Liu JR, Vaziri ND, Guo Y, Zhao YY. Microbiome-metabolome reveals the contribution of gut-kidney axis on kidney disease. J Transl Med 2019; 17:5. [PMID: 30602367 PMCID: PMC6317198 DOI: 10.1186/s12967-018-1756-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
Dysbiosis represents changes in composition and structure of the gut microbiome community (microbiome), which may dictate the physiological phenotype (health or disease). Recent technological advances and efforts in metagenomic and metabolomic analyses have led to a dramatical growth in our understanding of microbiome, but still, the mechanisms underlying gut microbiome–host interactions in healthy or diseased state remain elusive and their elucidation is in infancy. Disruption of the normal gut microbiota may lead to intestinal dysbiosis, intestinal barrier dysfunction, and bacterial translocation. Excessive uremic toxins are produced as a result of gut microbiota alteration, including indoxyl sulphate, p-cresyl sulphate, and trimethylamine-N-oxide, all implicated in the variant processes of kidney diseases development. This review focuses on the pathogenic association between gut microbiota and kidney diseases (the gut–kidney axis), covering CKD, IgA nephropathy, nephrolithiasis, hypertension, acute kidney injury, hemodialysis and peritoneal dialysis in clinic. Targeted interventions including probiotic, prebiotic and symbiotic measures are discussed for their potential of re-establishing symbiosis, and more effective strategies for the treatment of kidney diseases patients are suggested. The novel insights into the dysbiosis of the gut microbiota in kidney diseases are helpful to develop novel therapeutic strategies for preventing or attenuating kidney diseases and complications.![]()
Collapse
Affiliation(s)
- Yuan-Yuan Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Dan-Qian Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Lin Chen
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Jing-Ru Liu
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, CA, 92897, USA
| | - Yan Guo
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China.,Department of Internal Medicine, University of New Mexico, Albuquerque, 87131, USA
| | - Ying-Yong Zhao
- School of Pharmacy, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, Shaanxi, China.
| |
Collapse
|
40
|
Wang XQ, Zhang AH, Miao JH, Sun H, Yan GL, Wu FF, Wang XJ. Gut microbiota as important modulator of metabolism in health and disease. RSC Adv 2018; 8:42380-42389. [PMID: 35558413 PMCID: PMC9092240 DOI: 10.1039/c8ra08094a] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/02/2018] [Indexed: 12/12/2022] Open
Abstract
The human gastrointestinal tract colonizes a large number of microbial microflora, forms a host-microbiota co-metabolism structure with the host to participate in various metabolic processes in the human body, and plays a major role in the host immune response. In addition, the dysbiosis of intestinal microbial homeostasis is closely related to many diseases. Thus, an in-depth understanding of the relationship between them is of importance for disease pathogenesis, prevention and treatment. The combined use of metagenomics, transcriptomics, proteomics and metabolomics techniques for the analysis of gut microbiota can reveal the relationship between microbiota and the host in many ways, which has become a hot topic of analysis in recent years. This review describes the mechanism of co-metabolites in host health, including short-chain fatty acids (SCFA) and bile acid metabolism. The metabolic role of gut microbiota in obesity, liver diseases, gastrointestinal diseases and other diseases is also summarized, and the research methods for multi-omics combined application on gut microbiota are summarized. According to the studies of the interaction mechanism between gut microbiota and the host, we have a better understanding of the use of intestinal microflora in the treatment of related diseases. It is hoped that the gut microbiota can be utilized to maintain human health, providing a reference for future research.
Collapse
Affiliation(s)
- Xiang-Qian Wang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant Nanning Guangxi China +86-451-82110818 +86-451-82110818
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China
| | - Ai-Hua Zhang
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China
| | - Jian-Hua Miao
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant Nanning Guangxi China +86-451-82110818 +86-451-82110818
| | - Hui Sun
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China
| | - Guang-Li Yan
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China
| | - Fang-Fang Wu
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant Nanning Guangxi China +86-451-82110818 +86-451-82110818
| | - Xi-Jun Wang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant Nanning Guangxi China +86-451-82110818 +86-451-82110818
- National Chinmedomics Research Center, Sino-America Chinmedomics Technology Collaboration Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin China
| |
Collapse
|
41
|
Crowley EK, Long-Smith CM, Murphy A, Patterson E, Murphy K, O'Gorman DM, Stanton C, Nolan YM. Dietary Supplementation with a Magnesium-Rich Marine Mineral Blend Enhances the Diversity of Gastrointestinal Microbiota. Mar Drugs 2018; 16:E216. [PMID: 29925774 PMCID: PMC6024889 DOI: 10.3390/md16060216] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/25/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence demonstrates that dietary supplementation with functional food ingredients play a role in systemic and brain health as well as in healthy ageing. Conversely, deficiencies in calcium and magnesium as a result of the increasing prevalence of a high fat/high sugar "Western diet" have been associated with health problems such as obesity, inflammatory bowel diseases, and cardiovascular diseases, as well as metabolic, immune, and psychiatric disorders. It is now recognized that modulating the diversity of gut microbiota, the population of intestinal bacteria, through dietary intervention can significantly impact upon gut health as well as systemic and brain health. In the current study, we show that supplementation with a seaweed and seawater-derived functional food ingredient rich in bioactive calcium and magnesium (0.1% supplementation) as well as 70 other trace elements, significantly enhanced the gut microbial diversity in adult male rats. Given the significant impact of gut microbiota on health, these results position this marine multi-mineral blend (MMB) as a promising digestive-health promoting functional food ingredient.
Collapse
Affiliation(s)
- Erin K Crowley
- Department of Anatomy and Neuroscience, University College Cork, T12XF62 Cork, Ireland.
| | | | - Amy Murphy
- APC Microbiome Ireland, Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- School of Microbiology, University College Cork, T12YT20 Cork, Ireland.
| | - Elaine Patterson
- APC Microbiome Ireland, Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
| | - Kiera Murphy
- APC Microbiome Ireland, Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
| | - Denise M O'Gorman
- Marigot Ltd., Strand Farm, Currabinny, Carrigaline, P43NN62 Cork, Ireland.
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, University College Cork, T12YT20 Cork, Ireland.
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, T12XF62 Cork, Ireland.
- APC Microbiome Ireland, University College Cork, T12YT20 Cork, Ireland.
| |
Collapse
|