1
|
Ye P, Liu W, Tang X, Liu M, Han J, Wang X, Zhu J, He X, Zhu X, Cao M, Zhao L, Ren Q. Effects of hydroxypropyl starch on intestinal health and transcriptome of geese. Sci Rep 2025; 15:12284. [PMID: 40210970 PMCID: PMC11986093 DOI: 10.1038/s41598-025-96020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/25/2025] [Indexed: 04/12/2025] Open
Abstract
In recent years, gout resulting from uric acid metabolism disorders has led to significant economic losses in goose production. The intestine is a vital organ crucial for uric acid metabolism. Hydroxypropyl starch (HPS) is a resistant starch modified from natural starch, which can enhance intestinal health as a dietary ingredient fiber. In this study, 240 30-day-old Yangzhou geese with similar body weights were divided into three groups: The control group (CG) received a basal diet + 5% corn starch; the hydroxypropyl starch group (HPS) and the sodium urate group (SU) were given a basal diet + 5% hydroxypropyl starch. The experiment lasted for 21 days, and the SU group was administered 30 mg of sodium urate per day during the last 4 days of the study. The results indicated that the level of uric acid in the HPS group was 56.6 µmol/L, significantly lower than that in the CG group (70.8 µmol/L) and the SU group (129.7 µmol/L). The morphological findings revealed that the ileum of the CG group and the SU group exhibited varying degrees of damage, while the HPS group maintained complete structure. The villus height and the ratio of villus height to crypt depth in the HPS group were significantly higher compared to those in the CG and SU groups, while the crypt depth was significantly lower than that in the SU group. A total of 1462 differentially expressed genes (DEGs) were identified at the transcriptome level. GO and KEGG functional enrichment analyses indicated that the DEGs were significantly enriched in the Brush border membrane, Brush border, PPAR signaling pathway, PI3K-Akt signaling pathway, and other related processes. Subsequent analysis revealed that HPS up-regulated the expression of genes associated with intestinal function (such as SLC5A12 and SLC5A8), structure (including NR5A2, IPMK), and uric acid metabolism (PDZK1). The accuracy and reliability of transcriptome sequencing data were confirmed by RT-qPCR. In this study, we systematically demonstrated that HPS can improve intestinal morphology and reduce serum uric acid levels, emphasizing its potential as a dietary supplement for geese.
Collapse
Affiliation(s)
- Pengfei Ye
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China.
| | - Wenquan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xiaotong Tang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Mengxue Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Jingfan Han
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xiaoxue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Jie Zhu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xiaorong He
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xueqi Zhu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Mixia Cao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Lei Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Qingchang Ren
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China.
| |
Collapse
|
2
|
Yu J, Li Y, Zhu B, Shen J, Miao L. Research progress on the kidney-gut-brain axis in brain dysfunction in maintenance hemodialysis patients. Front Med (Lausanne) 2025; 12:1538048. [PMID: 40115780 PMCID: PMC11922870 DOI: 10.3389/fmed.2025.1538048] [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: 12/02/2024] [Accepted: 02/14/2025] [Indexed: 03/23/2025] Open
Abstract
Maintenance hemodialysis (MHD) has become the primary renal replacement therapy for patients with end-stage renal disease. The kidney-gut-brain axis represents a communication network connecting the kidney, intestine and brain. In MHD patients, factors such as uremic toxins, hemodynamic changes, vascular damage, inflammation, oxidative stress, and intestinal dysbiosis in MHD patients refers to a range of clinical syndromes, including brain injury, and is manifested by conditions such as white matter disease, brain atrophy, cerebrovascular disease, cognitive impairment, depression, anxiety, and other behavioral or consciousness abnormalities. Numerous studies have demonstrated the prevalence of these brain disorders in MHD patients. Understanding the mechanisms of brain disorders in MHD patients, particularly through the lens of kidney-gut-brain axis dysfunction, offers valuable insights for future research and the development of targeted therapies. This article reviews the brain dysfunction associated with MHD, the impact of the kidney-brain axis, intestinal barrier damage, gut microbiota dysbiosis caused by MHD, and the role of the gut-brain axis in brain dysfunction.
Collapse
Affiliation(s)
- Jie Yu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yulu Li
- Department of Nephrology, Taicang Loujiang New City Hospital, Suzhou, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jianqin Shen
- Department of Blood Purification Center, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liying Miao
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
3
|
Headley SA, Chapman DJ, Germain MJ, Evans EE, Madsen KL, Miele EM, Kirton K, Loseke J, Cornelius A, Martin B, Nindl B, Park H, Vaziri ND, Ikizler TA. Effects of High Amylose-Resistant Starch on Gut Microbiota and Uremic Toxin Levels in Patients With Stage-G3a-G4 Chronic Kidney Disease: A Randomized Trial. J Ren Nutr 2025; 35:248-258. [PMID: 39362281 DOI: 10.1053/j.jrn.2024.09.005] [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: 03/26/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVE This study was designed to determine the effect of 16 weeks of supplementation with Hi-maize 260 resistant starch (RS) on the gut microbiota, uremic toxins (indoxyl sulfate and p-cresyl sulfate [PCS]), markers of inflammation, and oxidative stress along with vascular function in patients with stage G3a-G4 chronic kidney disease (CKD). DESIGN AND METHODS This was a double-blind, placebo-controlled, parallel-arm, randomized controlled trial. Sixty-eight patients with stage-G3a-G4 CKD were randomized to either RS with usual care or placebo and usual care. Patients attended four testing sessions as follows: two baseline (BL) visits and follow-up visits at 8 and 16 weeks. Fasting blood samples, resting brachial and central blood pressures, along with arterial stiffness, were collected at visits (1 or 2) and weeks 8 and 16. A stool sample was collected for analysis of microbial composition at BL and week 16. Patients were randomized after the BL visits. RESULTS Patients receiving the RS had a reduction in PCS at week 16. This reduction was associated with a decrease in microbial α-diversity between BL and week 16 (Chao1 P = .014, Shannon P = .017, phylogenetic diversity P = .046, and Simpson P = .017) as well as increases in Subdoligranulum (P = .03) and Oscillospiraceae Unclassified Clostridiales Group 002 (P = .02) and decreases in Bacteroides (P = .009).There were no changes in microbial beta diversity and other biomarkers or markers of vascular function following the 16-week period. CONCLUSION Sixteen weeks of supplementation of RS in patients with stage-G3a-G4 CKD led to changes in microbial composition that were associated with a significant reduction in PCS.
Collapse
Affiliation(s)
- Samuel A Headley
- Department of Exercise Science, Springfield College, Springfield, Massachusetts.
| | - Donna J Chapman
- Department of Exercise Science, Springfield College, Springfield, Massachusetts
| | - Michael J Germain
- Renal and Transplant Associates of New England, Springfield, Massachusetts
| | - Elizabeth E Evans
- Department of Exercise Science, Springfield College, Springfield, Massachusetts
| | - Karen L Madsen
- Department of Gastroenterology, University of Alberta, Edmonton, Canada
| | - Emily M Miele
- Department of Exercise Science, Springfield College, Springfield, Massachusetts
| | - Kristyn Kirton
- Department of Exercise Science, Springfield College, Springfield, Massachusetts
| | - Joshua Loseke
- Department of Exercise Science, Springfield College, Springfield, Massachusetts
| | - Allen Cornelius
- School of Psychology, Fielding Graduate University, Colorado Springs, Colorado
| | - Brian Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bradley Nindl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Heekuk Park
- Division of Infectious Diseases, Department of Medicine, Microbiome & Pathogen Genomics Core Columbia University Medical Center, New York, New York
| | - Nosratola D Vaziri
- Emeritus Professor of Medicine, Physiology and Biophysics School of Medicine, University of California Irvine, Irvine, California
| | - Talat Alp Ikizler
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
4
|
Hayeeawaema F, Sermwittayawong N, Tipbunjong C, Huipao N, Muangnil P, Khuituan P. Live and heat-killed Leuconostoc mesenteroides counteract the gastrointestinal dysfunction in chronic kidney disease mice through intestinal environment modulation. PLoS One 2025; 20:e0318827. [PMID: 39992980 PMCID: PMC12005673 DOI: 10.1371/journal.pone.0318827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Probiotics are well-known therapeutic agents for managing constipation and have been used to improve chronic kidney disease (CKD) progression. However, heat-killed probiotics on CKD remain inadequately explored. This study aimed to evaluate the probiotic potential of lactic acid bacteria derived from natural sources and to investigate the effects of both live and heat-killed Leuconostoc mesenteroides (Ln.m) on renal and gastrointestinal functions in CKD mice. Ln.m was selected from acid and bile salt intolerance tests, non-hemolytic activity, and antibiotic sensitivity. CKD mice demonstrated significantly elevated blood urea nitrogen (BUN) and creatinine levels compared to control mice (p < 0.001 and p < 0.01). Treatment with live and heat-killed Ln.m significantly reduced BUN and creatinine levels in CKD mice (p < 0.01 and p < 0.05). Additionally, kidney damage observed in CKD mice compared to control mice, including glomerular necrosis, tubular dilatation, inflammation, and fibrosis, was significantly alleviated following live and heat-killed Ln.m treatments. CKD-induced gastrointestinal dysfunction was characterized by an imbalance in Firmicutes/Bacteroidota populations, increased colonic uremic toxin (p < 0.01), reduced fecal short-chain fatty acids (SCFAs) (p < 0.05), and constipation. Treatment with live and heat-killed Ln.m restored gut microbiota, decreased uremic toxin (p < 0.001), increased SCFAs (p < 0.05), and alleviated constipation. In summary, both live and heat-killed Ln.m effectively alleviated gastrointestinal dysfunction and renal damage in CKD mice, primarily through modulation of the intestinal environment. These findings highlight the therapeutic potential of live and heat-killed Ln.m as the gastrointestinal dysfunction treatment in CKD.
Collapse
Affiliation(s)
- Fittree Hayeeawaema
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Thailand
| | | | - Chittipong Tipbunjong
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Thailand
| | - Nawiya Huipao
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Thailand
| | - Paradorn Muangnil
- Faculty of Veterinary Science, Prince of Songkla University, Hat Yai, Thailand
| | - Pissared Khuituan
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Thailand
| |
Collapse
|
5
|
Han J, Zhang H, Li X, Tang Y, Du Y, Zhang H, Liao D. Relationship between dietary consumption of live microbes with mortality in adults with chronic kidney disease. J Nephrol 2025:10.1007/s40620-025-02212-w. [PMID: 39939503 DOI: 10.1007/s40620-025-02212-w] [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: 04/26/2024] [Accepted: 01/02/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND The connection between gut dysbiosis and chronic kidney disease (CKD) has been recognized, but, the effect of dietary intake of live microbes on the prognosis of CKD is still unclear. This analysis examined the relationship of dietary live microbe intake with mortality among adults with CKD. METHODS For this study, information was gathered from the National Health and Nutrition Examination Survey 1999-2018, which included 8725 adult participants with CKD. MedHi refers to the live microbial content of food beyond 104 CFU/g. To elucidate the link between MedHi dietary live microbe intake and mortality from all-cause and cardiovascular disease (CVD), we implemented a weighted multivariate Cox regression analysis. RESULTS In contrast to survivors, non-survivors had a lower intake of dietary live microbes. The findings from the multivariable model indicated a negative and linear relationship between an increment of 100 g in MedHi foods and reduced mortality from all-causes and CVD. Likewise, participants in the highest MedHi food group exhibited a 20% and 26% decreased risk of all-cause and CVD mortality, respectively, compared to those in the lowest MedHi food group. Stratified analyses conducted on various subgroups yielded consistent findings. CONCLUSION A significant inverse linear relationship was found between high dietary live microbe consumption and reduced all-cause and CVD mortality.
Collapse
Affiliation(s)
- Jianxin Han
- Department of Nephrology, The People's Hospital of Renshou County, The Second People's Hospital of Meishan City, Meishan, China
| | - Huan Zhang
- North Sichuan Medical College, Nanchong, China
| | - Xinchun Li
- North Sichuan Medical College, Nanchong, China
| | - Yumei Tang
- Department of Nephrology, The People's Hospital of Renshou County, The Second People's Hospital of Meishan City, Meishan, China
| | - Yunfei Du
- Chengdu Medical College, Chengdu, China
| | - Haiyan Zhang
- Pingshan County Hospital of Chinese Medicine, Yibin, Sichuan, China
| | - Dan Liao
- Department of Nephrology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China.
| |
Collapse
|
6
|
Zheng G, Cao J, Wang XH, He W, Wang B. The gut microbiome, chronic kidney disease, and sarcopenia. Cell Commun Signal 2024; 22:558. [PMID: 39574190 PMCID: PMC11580515 DOI: 10.1186/s12964-024-01922-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/01/2024] [Indexed: 11/25/2024] Open
Abstract
Sarcopenia is a prevalent condition in patients with chronic kidney disease (CKD), intricately linked to adverse prognoses, heightened cardiovascular risks, and increased mortality rates. Extensive studies have found a close and complex association between gut microbiota, kidney and muscle. On one front, patients with CKD manifest disturbances in gut microbiota and alterations in serum metabolites. These abnormal microbiota composition and metabolites in turn participate in the development of CKD. On another front, altered gut microbiota and its metabolites may lead to significant changes in metabolic homeostasis and inflammation, ultimately contributing to the onset of sarcopenia. The disturbance of gut microbial homeostasis, coupled with the accumulation of toxic metabolites, exerts deleterious effects on skeletal muscles in CKD patients with sarcopenia. This review meticulously describes the alterations observed in gut microbiota and its serum metabolites in CKD and sarcopenia patients, providing a comprehensive overview of pertinent studies. By delving into the intricate interplay of gut microbiota and serum metabolites in CKD-associated sarcopenia, we aim to unveil novel treatment strategies for ameliorating their symptoms and prognosis.
Collapse
Affiliation(s)
- Guohao Zheng
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Jingyuan Cao
- Institute of Nephrology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Xiaonan H Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Wei He
- Department of Gastroenterology, Geriatric Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
7
|
Coll E, Cigarran S, Portolés J, Cases A. Gut Dysbiosis and Its Role in the Anemia of Chronic Kidney Disease. Toxins (Basel) 2024; 16:495. [PMID: 39591250 PMCID: PMC11598790 DOI: 10.3390/toxins16110495] [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: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The gut dysbiosis present in chronic kidney disease (CKD) has been associated with anemia. Factors such as the accumulation of gut-derived uremic toxins, increased gut barrier permeability-induced inflammation, and a reduced intestinal production of short-chain fatty acids (SCFAs), all associated with changes in the intestinal microbiota composition in CKD, may lead to the development or worsening of anemia in renal patients. Understanding and addressing these mechanisms related to gut dysbiosis in CKD patients can help to delay the development of anemia and improve its control in this population. One approach is to avoid or reduce the use of drugs linked to gut dysbiosis in CKD, such as phosphate binders, oral iron supplementation, antibiotics, and others, unless they are indispensable. Another approach involves introducing dietary changes that promote a healthier microbiota and/or using prebiotics, probiotics, or symbiotics to improve gut dysbiosis in this setting. These measures can increase the presence of SCFA-producing saccharolytic bacteria and reduce proteolytic bacteria, thereby lowering the production of gut-derived uremic toxins and inflammation. By ameliorating CKD-related gut dysbiosis, these strategies can also improve the control of renal anemia and enhance the response to erythropoiesis-stimulating agents (ESAs) in ESA-resistant patients. In this review, we have explored the relationship between gut dysbiosis in CKD and renal anemia and propose feasible solutions, both those already known and potential future treatments.
Collapse
Affiliation(s)
- Elisabet Coll
- Servei de Nefrologia, Fundacio Puigvert, 08025 Barcelona, Spain
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
| | | | - Jose Portolés
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
- Ressearch Net RICORS 2030 Instituto de Salud Carlos III ISCIII, 28029 Madrid, Spain
- Nephrology Department, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain
- Medicine Department, Facultad de Medicina, Research Institute Puerta de Hierro Segovia de Arana (IDIPHISA), Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Aleix Cases
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
- Nephrology Unit, Hospital Clinic, 08036 Barcelona, Spain
| |
Collapse
|
8
|
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
|
9
|
Papetti L, Del Chierico F, Frattale I, Toto F, Scanu M, Mortera SL, Rapisarda F, Di Michele M, Monte G, Ursitti F, Sforza G, Putignani L, Valeriani M. Pediatric migraine is characterized by traits of ecological and metabolic dysbiosis and inflammation. J Headache Pain 2024; 25:171. [PMID: 39379796 PMCID: PMC11462686 DOI: 10.1186/s10194-024-01871-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Recently, there has been increasing interest in the possible role of the gut microbiota (GM) in the onset of migraine. Our aim was to verify whether bacterial populations associated with intestinal dysbiosis are found in pediatric patients with migraine. We looked for which metabolic pathways, these bacteria were involved and whether they might be associated with gut inflammation and increased intestinal permeability. METHODS Patients aged between 6 and 17 years were recruited. The GM profiling was performed by the 16S rRNA metataxonomics of faecal samples from 98 patients with migraine and 98 healthy subjects. Alpha and beta diversity analyses and multivariate and univariate analyses were applied to compare the gut microbiota profiles between the two group. To predict functional metabolic pathways, we used phylogenetic analysis of communities. The level of indican in urine was analyzed to investigate the presence of metabolic dysbiosis. To assess gut inflammation, increased intestinal permeability and the mucosal immune activation, we measured the plasmatic levels of lipopolysaccharide, occludin and IgA, respectively. RESULTS The α-diversity analysis revealed a significant increase of bacterial richness in the migraine group. The β-diversity analysis showed significant differences between the two groups indicating gut dysbiosis in patients with migraine. Thirty-seven metabolic pathways were increased in the migraine group, which includes changes in tryptophan and phenylalanine metabolism. The presence of metabolic dysbiosis was confirmed by the increased level of indican in urine. Increased levels of plasmatic occludin and IgA indicated the presence of intestinal permeability and mucosal immune activation. The plasmatic LPS levels showed a low intestinal inflammation in patients with migraine. CONCLUSIONS Pediatric patients with migraine present GM profiles different from healthy subjects, associated with metabolic pathways important in migraine.
Collapse
Affiliation(s)
- Laura Papetti
- Developmental Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio, 4, Rome, Italy
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy.
| | - Ilaria Frattale
- Child Neurology and Psychiatry Unit, Department of Wellbeing of Mental and Neurological, Dental and Sensory Organ Health, Policlinico Tor Vergata Foundation Hospital, Rome, Italy
| | - Francesca Toto
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy
| | - Stefano Levi Mortera
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy
| | - Federica Rapisarda
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy
| | - Marta Di Michele
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy
| | - Gabriele Monte
- Developmental Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio, 4, Rome, Italy
| | - Fabiana Ursitti
- Developmental Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio, 4, Rome, Italy
| | - Giorgia Sforza
- Developmental Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio, 4, Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Research Area of Immunology, Rheumatology and Infectious Diseases, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Viale Di San Paolo, 15, Rome, Italy.
| | - Massimiliano Valeriani
- Developmental Neurology Unit, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio, 4, Rome, Italy.
- Systems Medicine Department, Tor Vergata University of Rome, Rome, Italy.
- Center for Sensory-Motor Interaction, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
10
|
Guo M, He F, Zhang C. Molecular Therapeutics for Diabetic Kidney Disease: An Update. Int J Mol Sci 2024; 25:10051. [PMID: 39337537 PMCID: PMC11431964 DOI: 10.3390/ijms251810051] [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: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication of diabetes mellitus (DM). With the increasing prevalence of DM worldwide, the incidence of DKD remains high. If DKD is not well controlled, it can develop into chronic kidney disease or end-stage renal disease (ESRD), which places considerable economic pressure on society. Traditional therapies, including glycemic control, blood pressure control, blood lipid control, the use of renin-angiotensin system blockers and novel drugs, such as sodium-glucose cotransporter 2 inhibitors, mineralocorticoid receptor inhibitors and glucagon-like peptide-1 receptor agonists, have been used in DKD patients. Although the above treatment strategies can delay the progression of DKD, most DKD patients still ultimately progress to ESRD. Therefore, new and multimodal treatment methods need to be explored. In recent years, researchers have continuously developed new treatment methods and targets to delay the progression of DKD, including miRNA therapy, stem cell therapy, gene therapy, gut microbiota-targeted therapy and lifestyle intervention. These new molecular therapy methods constitute opportunities to better understand and treat DKD. In this review, we summarize the progress of molecular therapeutics for DKD, leading to new treatment strategies.
Collapse
Affiliation(s)
| | - Fangfang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
11
|
Liabeuf S, Hafez G, Pešić V, Spasovski G, Bobot M, Mačiulaitis R, Bumblyte IA, Ferreira AC, Farinha A, Malyszko J, Pépin M, Massy ZA, Unwin R, Capasso G, Mani LY. Drugs with a negative impact on cognitive functions (part 3): antibacterial agents in patients with chronic kidney disease. Clin Kidney J 2024; 17:sfae174. [PMID: 39114495 PMCID: PMC11304602 DOI: 10.1093/ckj/sfae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Indexed: 08/10/2024] Open
Abstract
The relationship between chronic kidney disease (CKD) and cognitive function has received increased attention in recent years. Antibacterial agents (ABs) represent a critical component of therapy regimens in patients with CKD due to increased susceptibility to infections. Following our reviewing work on the neurocognitive impact of long-term medications in patients with CKD, we propose to focus on AB-induced direct and indirect consequences on cognitive function. Patients with CKD are predisposed to adverse drug reactions (ADRs) due to altered drug pharmacokinetics, glomerular filtration decline, and the potential disruption of the blood-brain barrier. ABs have been identified as a major cause of ADRs in vulnerable patient populations. This review examines the direct neurotoxic effects of AB classes (e.g. beta-lactams, fluoroquinolones, aminoglycosides, and metronidazole) on the central nervous system (CNS) in patients with CKD. We will mainly focus on the acute effects on the CNS associated with AB since they are the most extensively studied effects in CKD patients. Moreover, the review describes the modulation of the gut microbiota by ABs, potentially influencing CNS symptoms. The intricate brain-gut-kidney axis emerges as a pivotal focus, revealing the interplay between microbiota alterations induced by ABs and CNS manifestations in patients with CKD. The prevalence of antibiotic-associated encephalopathy in patients with CKD undergoing intravenous AB therapy supports the use of therapeutic drug monitoring for ABs to reduce the number and seriousness of ADRs in this patient population. In conclusion, elucidating AB-induced cognitive effects in patients with CKD demands a comprehensive understanding and tailored therapeutic strategies that account for altered pharmacokinetics and the brain-gut-kidney axis.
Collapse
Affiliation(s)
- Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens University Medical Center, Amiens, France
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, Amiens, France
| | - Gaye Hafez
- Department of Pharmacology, Faculty of Pharmacy, Altinbas University, Istanbul, Turkey
| | - Vesna Pešić
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Goce Spasovski
- Department of Nephrology, Clinical Centre “Mother Theresa”, Saints Cyril and Methodius University, Skopje, North Macedonia
| | - Mickaël Bobot
- Aix-Marseille University, Department of Nephrology, AP-HM, La Conception Hospital, Marseille, France; C2VN Laboratory, Inserm 1263, INRAE 1260, Aix-Marseille University, Marseille, France
| | - Romaldas Mačiulaitis
- Department of Nephrology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Institute of Physiology and Pharmacology, Faculty of Medicines, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ana Carina Ferreira
- Nephrology Department, Centro Hospitalar e Universitário de Lisboa Central, Lisbon, Portugal
- Universidade Nova de Lisboa-Faculdade de Ciências Médicas-Nephology, Lisbon, Portugal
| | - Ana Farinha
- Department of Nephrology, Hospital de Vila Franca de Xira, Lisbon, Portugal
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marion Pépin
- Department of Geriatrics, Ambroise Paré University Medical Center, APHP, Boulogne-Billancourt, France
- Paris-Saclay University, UVSQ, Inserm, Clinical Epidemiology Team, Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
| | - Ziad A Massy
- Paris-Saclay University, UVSQ, Inserm, Clinical Epidemiology Team, Centre de Recherche en Epidémiologie et Santé des Populations (CESP), Villejuif, France
- Department of Nephrology, Ambroise Paré University Medical Center, APHP, Paris, France
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Giovambattista Capasso
- Department of Translantional Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
- Biogem Research Institute, Ariano Irpino, Italy
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
12
|
Anwer M, Wei MQ. Harnessing the power of probiotic strains in functional foods: nutritive, therapeutic, and next-generation challenges. Food Sci Biotechnol 2024; 33:2081-2095. [PMID: 39130669 PMCID: PMC11315846 DOI: 10.1007/s10068-024-01630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 08/13/2024] Open
Abstract
Functional foods have become an essential element of the diet in developed nations, due to their health benefits and nutritive values. Such food products are only called functional if they, "In addition to basic nutrition, have valuable effects on one or multiple functions of the human body, thereby enhancing general and physical conditions and/or reducing the risk of disease progression". Functional foods are currently one of the most extensively researched areas in the food and nutrition sciences. They are fortified and improved food products. Presently, probiotics are regarded as the most significant and commonly used functional food product. Diverse probiotic food products and supplements are used according to the evidence that supports their strength, functionality, and recommended dosage. This review provides an overview of the current functional food market, with a particular focus on probiotic microorganisms as pivotal functional ingredients. It offers insights into current research endeavors and outlines potential future directions in the field.
Collapse
Affiliation(s)
- Muneera Anwer
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Gold Coast Campus, Parklands Drive, Southport, QLD 4215 Australia
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Ming Q. Wei
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Gold Coast Campus, Parklands Drive, Southport, QLD 4215 Australia
| |
Collapse
|
13
|
Cladis DP, Burstad KM, Biruete A, Jannasch AH, Cooper BR, Hill Gallant KM. Dietary Phosphorus Levels Influence Protein-Derived Uremic Toxin Production in Nephrectomized Male Rats. Nutrients 2024; 16:1807. [PMID: 38931160 PMCID: PMC11207110 DOI: 10.3390/nu16121807] [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: 05/20/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Gut microbiota-derived uremic toxins (UT) accumulate in patients with chronic kidney disease (CKD). Dietary phosphorus and protein restriction are common in CKD treatment, but the relationship between dietary phosphorus, a key nutrient for the gut microbiota, and protein-derived UT is poorly studied. Thus, we explored the relationship between dietary phosphorus and serum UT in CKD rats. For this exploratory study, we used serum samples from a larger study on the effects of dietary phosphorus on intestinal phosphorus absorption in nephrectomized (Nx, n = 22) or sham-operated (sham, n = 18) male Sprague Dawley rats. Rats were randomized to diet treatment groups of low or high phosphorus (0.1% or 1.2% w/w, respectively) for 1 week, with serum trimethylamine oxide (TMAO), indoxyl sulfate (IS), and p-cresol sulfate (pCS) analyzed by LC-MS. Nx rats had significantly higher levels of serum TMAO, IS, and pCS compared to sham rats (all p < 0.0001). IS showed a significant interaction between diet and CKD status, where serum IS was higher with the high-phosphorus diet in both Nx and sham rats, but to a greater extent in the Nx rats. Serum TMAO (p = 0.24) and pCS (p = 0.34) were not affected by dietary phosphorus levels. High dietary phosphorus intake for 1 week results in higher serum IS in both Nx and sham rats. The results of this exploratory study indicate that reducing dietary phosphorus intake in CKD may have beneficial effects on UT accumulation.
Collapse
Affiliation(s)
- Dennis P. Cladis
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (D.P.C.); (K.M.B.)
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA;
- Department of Food Science and Technology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Kendal M. Burstad
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (D.P.C.); (K.M.B.)
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA;
| | - Annabel Biruete
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA;
- Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amber H. Jannasch
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47906, USA; (A.H.J.); (B.R.C.)
| | - Bruce R. Cooper
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47906, USA; (A.H.J.); (B.R.C.)
| | - Kathleen M. Hill Gallant
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN 55108, USA; (D.P.C.); (K.M.B.)
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA;
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
Vandecruys M, De Smet S, De Beir J, Renier M, Leunis S, Van Criekinge H, Glorieux G, Raes J, Vanden Wyngaert K, Nagler E, Calders P, Monbaliu D, Cornelissen V, Evenepoel P, Van Craenenbroeck AH. Revitalizing the Gut Microbiome in Chronic Kidney Disease: A Comprehensive Exploration of the Therapeutic Potential of Physical Activity. Toxins (Basel) 2024; 16:242. [PMID: 38922137 PMCID: PMC11209503 DOI: 10.3390/toxins16060242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Both physical inactivity and disruptions in the gut microbiome appear to be prevalent in patients with chronic kidney disease (CKD). Engaging in physical activity could present a novel nonpharmacological strategy for enhancing the gut microbiome and mitigating the adverse effects associated with microbial dysbiosis in individuals with CKD. This narrative review explores the underlying mechanisms through which physical activity may favorably modulate microbial health, either through direct impact on the gut or through interorgan crosstalk. Also, the development of microbial dysbiosis and its interplay with physical inactivity in patients with CKD are discussed. Mechanisms and interventions through which physical activity may restore gut homeostasis in individuals with CKD are explored.
Collapse
Affiliation(s)
- Marieke Vandecruys
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
| | - Stefan De Smet
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, 3000 Leuven, Belgium;
| | - Jasmine De Beir
- Department of Rehabilitation Sciences, Ghent University, 9000 Ghent, Belgium; (J.D.B.); (P.C.)
| | - Marie Renier
- Group Rehabilitation for Internal Disorders, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (M.R.); (V.C.)
| | - Sofie Leunis
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
| | - Hanne Van Criekinge
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
| | - Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Jeroen Raes
- Department of Microbiology and Immunology, Rega Institute for Medical Research, 3000 Leuven, Belgium;
- VIB-KU Leuven Center for Microbiology, 3000 Leuven, Belgium
| | - Karsten Vanden Wyngaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Evi Nagler
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, 9000 Ghent, Belgium; (G.G.); (K.V.W.); (E.N.)
| | - Patrick Calders
- Department of Rehabilitation Sciences, Ghent University, 9000 Ghent, Belgium; (J.D.B.); (P.C.)
| | - Diethard Monbaliu
- Department of Microbiology, Immunology and Transplantation, Abdominal Transplantation, KU Leuven, 3000 Leuven, Belgium; (S.L.); (H.V.C.); (D.M.)
- Transplantoux Foundation, 3000 Leuven, Belgium
| | - Véronique Cornelissen
- Group Rehabilitation for Internal Disorders, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium; (M.R.); (V.C.)
| | - Pieter Evenepoel
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
- Department of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Amaryllis H. Van Craenenbroeck
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium; (M.V.); or (P.E.)
- Department of Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
15
|
Pires L, González-Paramás AM, Heleno SA, Calhelha RC. The Role of Gut Microbiota in the Etiopathogenesis of Multiple Chronic Diseases. Antibiotics (Basel) 2024; 13:392. [PMID: 38786121 PMCID: PMC11117238 DOI: 10.3390/antibiotics13050392] [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: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic diseases (CD) may result from a combination of genetic factors, lifestyle and social behaviours, healthcare system influences, community factors, and environmental determinants of health. These risk factors frequently coexist and interact with one another. Ongoing research and a focus on personalized interventions are pivotal strategies for preventing and managing chronic disease outcomes. A wealth of literature suggests the potential involvement of gut microbiota in influencing host metabolism, thereby impacting various risk factors associated with chronic diseases. Dysbiosis, the perturbation of the composition and activity of the gut microbiota, is crucial in the etiopathogenesis of multiple CD. Recent studies indicate that specific microorganism-derived metabolites, including trimethylamine N-oxide, lipopolysaccharide and uremic toxins, contribute to subclinical inflammatory processes implicated in CD. Various factors, including diet, lifestyle, and medications, can alter the taxonomic species or abundance of gut microbiota. Researchers are currently dedicating efforts to understanding how the natural progression of microbiome development in humans affects health outcomes. Simultaneously, there is a focus on enhancing the understanding of microbiome-host molecular interactions. These endeavours ultimately aim to devise practical approaches for rehabilitating dysregulated human microbial ecosystems, intending to restore health and prevent diseases. This review investigates how the gut microbiome contributes to CD and explains ways to modulate it for managing or preventing chronic conditions.
Collapse
Affiliation(s)
- Lara Pires
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (S.A.H.)
- Grupo de Investigación en Polifenoles en Alimentos, Implicaciones en la Calidad y en Salud Humana, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain;
| | - Ana M. González-Paramás
- Grupo de Investigación en Polifenoles en Alimentos, Implicaciones en la Calidad y en Salud Humana, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain;
| | - Sandrina A. Heleno
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (S.A.H.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Ricardo C. Calhelha
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (S.A.H.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| |
Collapse
|
16
|
Krishnamoorthy N, Kalyan M, Hediyal TA, Anand N, Kendaganna PH, Pendyala G, Yelamanchili SV, Yang J, Chidambaram SB, Sakharkar MK, Mahalakshmi AM. Role of the Gut Bacteria-Derived Metabolite Phenylacetylglutamine in Health and Diseases. ACS OMEGA 2024; 9:3164-3172. [PMID: 38284070 PMCID: PMC10809373 DOI: 10.1021/acsomega.3c08184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024]
Abstract
Over the past few decades, it has been well established that gut microbiota-derived metabolites can disrupt gut function, thus resulting in an array of diseases. Notably, phenylacetylglutamine (PAGln), a bacterial derived metabolite, has recently gained attention due to its role in the initiation and progression of cardiovascular and cerebrovascular diseases. This meta-organismal metabolite PAGln is a byproduct of amino acid acetylation of its precursor phenylacetic acid (PAA) from a range of dietary sources like egg, meat, dairy products, etc. The microbiota-dependent metabolism of phenylalanine produces PAA, which is a crucial intermediate that is catalyzed by diverse microbial catalytic pathways. PAA conjugates with glutamine and glycine in the liver and kidney to predominantly form phenylacetylglutamine in humans and phenylacetylglycine in rodents. PAGln is associated with thrombosis as it enhances platelet activation mediated through the GPCRs receptors α2A, α2B, and β2 ADRs, thereby aggravating the pathological conditions. Clinical evidence suggests that elevated levels of PAGln are associated with pathology of cardiovascular, cerebrovascular, and neurological diseases. This Review further consolidates the microbial/biochemical synthesis of PAGln and discusses its role in the above pathophysiologies.
Collapse
Affiliation(s)
- Naveen
Kumar Krishnamoorthy
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Manjunath Kalyan
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Tousif Ahmed Hediyal
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Nikhilesh Anand
- Department
of Pharmacology, College of Medicine, American
University of Antigua, P. O. Box W-1451, Saint John’s, Antigua and Barbuda
| | - Pavan Heggadadevanakote Kendaganna
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Gurudutt Pendyala
- Department
of Anesthesiology, University of Nebraska
Medical Center (UNMC), Omaha, Nebraska 68198, United States
- Department
of Genetics, Cell Biology, and Anatomy, UNMC, Omaha, Nebraska 68198, United States
- Child Health
Research Institute, UNMC, Omaha, Nebraska 68198, United States
- National
Strategic Research Institute, UNMC, Omaha, Nebraska 68198, United States
| | - Sowmya V. Yelamanchili
- Department
of Anesthesiology, University of Nebraska
Medical Center (UNMC), Omaha, Nebraska 68198, United States
- Department
of Genetics, Cell Biology, and Anatomy, UNMC, Omaha, Nebraska 68198, United States
- National
Strategic Research Institute, UNMC, Omaha, Nebraska 68198, United States
| | - Jian Yang
- Drug
Discovery and Development Research Group, College of Pharmacy and
Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Saravana Babu Chidambaram
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Meena Kishore Sakharkar
- Drug
Discovery and Development Research Group, College of Pharmacy and
Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Arehally M. Mahalakshmi
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Centre
for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru 570015, India
| |
Collapse
|
17
|
Coutinho-Wolino KS, Melo MFS, Mota JC, Mafra D, Guimarães JT, Stockler-Pinto MB. Blueberry, cranberry, raspberry, and strawberry as modulators of the gut microbiota: target for treatment of gut dysbiosis in chronic kidney disease? From current evidence to future possibilities. Nutr Rev 2024; 82:248-261. [PMID: 37164634 DOI: 10.1093/nutrit/nuad048] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Gut dysbiosis is common in patients with chronic kidney disease (CKD) and is associated with uremic toxin production, inflammation, oxidative stress, and cardiovascular disease development. Therefore, healthy dietary patterns are essential modulators of gut microbiota. In this context, studies suggest that consuming berry fruits, rich in polyphenols and nutrients, may positively affect the gut microbiota, promoting the selective growth of beneficial bacteria and improving clinical status. However, studies on the effects of berry fruits on gut microbiota in CKD are scarce, and a better understanding of the possible mechanisms of action of berry fruits on gut microbiota is needed to guide future clinical studies and clinical practice in CKD. The objective was to discuss how berry fruits (blueberry, cranberry, raspberry, and strawberry) could be a therapeutic strategy to modulate the gut microbiota and possibly reverse the dysbiosis in CKD. Overall, available evidence shows that berry fruits can promote an increase in diversity by affecting the abundance of mucus-producing bacteria and short-chain fatty acids. Moreover, these fruits can increase the expression of mRNA involved in tight junctions in the gut such as occludin, tight junction protein 1 (TJP1), and mucin. Studies on the exact amount of berries leading to these effects show heterogeneous findings. However, it is known that, with 5 mg/day, it is already possible to observe some effects in animal models. Wild berries could possibly improve the uremic condition by reducing the levels of uremic toxins via modulation of the gut microbiota. In the long term, this could be an excellent strategy for patients with CKD. Therefore, clinical studies are encouraged to evaluate better these effects on CKD as well as the safe amount of these fruits in order to promote a better quality of life or even the survival of these patients.
Collapse
Affiliation(s)
- Karen S Coutinho-Wolino
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Manuela F S Melo
- Graduate Program in Nutrition, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
| | - Jessica C Mota
- Graduate Program in Nutrition, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
| | - Denise Mafra
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
- Postgraduate Program in Nutrition Sciences, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Medical Sciences, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Jonas T Guimarães
- Department of Food Technology, Faculty of Veterinary, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Milena B Stockler-Pinto
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
- Postgraduate Program in Nutrition Sciences, Faculty of Nutrition, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program in Pathology, Faculty of Medicine, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
18
|
Schwarz A, Hernandez L, Arefin S, Sartirana E, Witasp A, Wernerson A, Stenvinkel P, Kublickiene K. Sweet, bloody consumption - what we eat and how it affects vascular ageing, the BBB and kidney health in CKD. Gut Microbes 2024; 16:2341449. [PMID: 38686499 PMCID: PMC11062370 DOI: 10.1080/19490976.2024.2341449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
In today's industrialized society food consumption has changed immensely toward heightened red meat intake and use of artificial sweeteners instead of grains and vegetables or sugar, respectively. These dietary changes affect public health in general through an increased incidence of metabolic diseases like diabetes and obesity, with a further elevated risk for cardiorenal complications. Research shows that high red meat intake and artificial sweeteners ingestion can alter the microbial composition and further intestinal wall barrier permeability allowing increased transmission of uremic toxins like p-cresyl sulfate, indoxyl sulfate, trimethylamine n-oxide and phenylacetylglutamine into the blood stream causing an array of pathophysiological effects especially as a strain on the kidneys, since they are responsible for clearing out the toxins. In this review, we address how the burden of the Western diet affects the gut microbiome in altering the microbial composition and increasing the gut permeability for uremic toxins and the detrimental effects thereof on early vascular aging, the kidney per se and the blood-brain barrier, in addition to the potential implications for dietary changes/interventions to preserve the health issues related to chronic diseases in future.
Collapse
Affiliation(s)
- Angelina Schwarz
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Leah Hernandez
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Samsul Arefin
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Sartirana
- Department of Translational Medicine, Nephrology and Kidney Transplantation Unit, University of Piemonte Orientale, Novara, Italy
| | - Anna Witasp
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika Wernerson
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Karolina Kublickiene
- Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Ulrich C, Canim Z, Herberger E, Girndt M, Fiedler R. Inflammation in Hypervolemic Hemodialysis Patients: The Roles of RelB and Caspase-4. Int J Mol Sci 2023; 24:17550. [PMID: 38139378 PMCID: PMC10743509 DOI: 10.3390/ijms242417550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Hypervolemia is associated with inflammation in hemodialysis (HD) patients. How hypervolemia triggers inflammation is not entirely known. We initiated a cross-sectional study enrolling 40 hemodialysis patients who were categorized into normovolemic (N; 23) and hypervolemic (H; 17) groups by bioimpedance measurement. A caspase activity assay in combination with a specific caspase-4 inhibitor was used to detect caspase-4 activity in isolated peripheral blood mononuclear cells (PBMCs). Transcription factors RelA (pS529) and RelB (pS552) were analyzed by phospho-flow cytometry. Serum endotoxins were detected by an amebocyte lysate-based assay, and IL-6 (interleukin-6) and TNF-α (Tumor necrosis factor-α) gene expression were detected using the ELISA technique. Hypervolemic patients were older, more frequently had diabetes and showed increased CRP and IL-6 levels. Caspase-4 activity, which is linked to intracellular endotoxin detection, was significantly elevated in H patients. While the frequency of RelA-expressing immune cells and the expression density in these cells did not differ, the monocytic frequency of cells positively stained for RelB (pS552) was significantly decreased in H patients. Increased caspase-4 activity in H patients may indicate a cause of inflammation in H patients. The post-translational modification of RelB (pS552) is linked to downregulation of NF-kB activity and may indicate the resolution of inflammation, which is more distinct in N patients compared to H patients. Therefore, both higher inflammatory loads and lower inflammatory resolution capacities are characteristics of H patients.
Collapse
Affiliation(s)
- Christof Ulrich
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (Z.C.); (E.H.); (M.G.); (R.F.)
| | | | | | | | | |
Collapse
|
20
|
Mei Y, Tong X, Hu Y, Liu W, Wang J, Lv K, Li X, Cao L, Wang Z, Xiao W, Gao X. Comparative pharmacokinetics of six bioactive components of Shen-Wu-Yi-Shen tablets in normal and chronic renal failure rats based on UPLC-TSQ-MS/MS. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116818. [PMID: 37348793 DOI: 10.1016/j.jep.2023.116818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/12/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shen-Wu-Yi-Shen tablets (SWYST), a Chinese patent medicine consisting of 12 herbal medicines, was formulated by a famous TCM nephrologist, Zou Yunxiang. It is clinically used to improve the symptoms of nausea, vomiting, poor appetite, dry mouth and throat, and dry stool in patients with chronic renal failure (CRF) accompanied by qi and yin deficiency, dampness, and turbidity. SWYST can reduce urea nitrogen, blood creatinine, and urinary protein loss, and increase the endogenous creatinine clearance rate. However, little is known about its pharmacokinetics. AIM OF STUDY To compare the pharmacokinetics of six bioactive components after oral administration of SWYST in normal and adenine-induced CRF rats. MATERIALS AND METHODS A method based on ultra-performance liquid chromatography coupled with a triple-stage quadrupole mass spectrometer (UPLC-TSQ-MS/MS) was developed and validated to determine the six bioactive compounds (albiflorin, paeoniflorin, plantagoguanidinic acid, rhein, aloe-emodin, and emodin) in rat plasma. Rat plasma samples were prepared using protein precipitation. Chromatography was performed on an Agilent Eclipse Plus C18 column (3.0 × 50 mm, 1.8 μm) using gradient elution with a mobile phase composed of acetonitrile and water containing 0.1% (v/v) formic acid, while detection was achieved by electrospray ionization MS under the multiple selective reaction monitoring modes. After SWYST administration, rat plasma was collected at different time points, and the pharmacokinetic parameters of six analytes were calculated and analyzed based on the measured plasma concentrations. RESULTS The UPLC-TSQ-MS/MS method was fully validated for its satisfactory linearity (r ≥ 0.9913), good precisions (RSD <11.5%), and accuracy (RE: -13.4∼13.1%), as well as acceptable limits in the extraction recoveries, matrix effects, and stability (RSD <15%). In normal rats, the six analytes were rapidly absorbed (Tmax ≤ 2 h), and approximately 80% of their total exposure was eliminated within 10 h. Moreover, in normal rats, the AUC0-t and Cmax of albiflorin, plantagoguanidinic acid, and rhein exhibited linear pharmacokinetics within the dose ranges, while that of paeoniflorin is non-linear. However, in CRF rats, the six analytes exhibited reduced elimination and significantly different AUC or Cmax values. These changes may reflect a decreased renal clearance rate or inhibition of drug-metabolizing enzymes and transporters in the liver and gastrointestinal tract caused by CRF. CONCLUSIONS A sensitive UPLC-TSQ-MS/MS method was validated and used to investigate the pharmacokinetics of SWYST in normal and CRF rats. This is the first study to investigate the pharmacokinetics of SWYST, and our findings elucidate the causes of their different pharmacokinetic behaviors in CRF rats. Furthermore, the results provide useful information to guide further research on the pharmacokinetic-pharmacodynamic correlation and clinical application of SWYST.
Collapse
Affiliation(s)
- Yudan Mei
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Xiaoyu Tong
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Yumei Hu
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Wenjun Liu
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Jiajia Wang
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Kaihong Lv
- China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xu Li
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Liang Cao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Zhenzhong Wang
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China
| | - Wei Xiao
- College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China.
| | - Xia Gao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China; Local Joint Engineering Research Center on the Intelligent Manufacturing of TCM, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, 222047, People's Republic of China.
| |
Collapse
|
21
|
Altamura S, Pietropaoli D, Lombardi F, Del Pinto R, Ferri C. An Overview of Chronic Kidney Disease Pathophysiology: The Impact of Gut Dysbiosis and Oral Disease. Biomedicines 2023; 11:3033. [PMID: 38002033 PMCID: PMC10669155 DOI: 10.3390/biomedicines11113033] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Chronic kidney disease (CKD) is a severe condition and a significant public health issue worldwide, carrying the burden of an increased risk of cardiovascular events and mortality. The traditional factors that promote the onset and progression of CKD are cardiometabolic risk factors like hypertension and diabetes, but non-traditional contributors are escalating. Moreover, gut dysbiosis, inflammation, and an impaired immune response are emerging as crucial mechanisms in the disease pathology. The gut microbiome and kidney disease exert a reciprocal influence commonly referred to as "the gut-kidney axis" through the induction of metabolic, immunological, and endocrine alterations. Periodontal diseases are strictly involved in the gut-kidney axis for their impact on the gut microbiota composition and for the metabolic and immunological alterations occurring in and reciprocally affecting both conditions. This review aims to provide an overview of the dynamic biological interconnections between oral health status, gut, and renal pathophysiology, spotlighting the dynamic oral-gut-kidney axis and raising whether periodontal diseases and gut microbiota can be disease modifiers in CKD. By doing so, we try to offer new insights into therapeutic strategies that may enhance the clinical trajectory of CKD patients, ultimately advancing our quest for improved patient outcomes and well-being.
Collapse
Affiliation(s)
- Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- PhD School in Medicine and Public Health, Center of Oral Diseases, Prevention and Translational Research—Dental Clinic, 67100 L’Aquila, Italy
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
| | - Davide Pietropaoli
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
- Center of Oral Diseases, Prevention and Translational Research—Dental Clinic, 67100 L’Aquila, Italy
| | - Francesca Lombardi
- Laboratory of Immunology and Immunopathology, Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Rita Del Pinto
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
- Unit of Internal Medicine and Nephrology, Center for Hypertension and Cardiovascular Prevention, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.A.); (D.P.); (C.F.)
- Oral Diseases and Systemic Interactions Study Group (ODISSY Group), 67100 L’Aquila, Italy
- Unit of Internal Medicine and Nephrology, Center for Hypertension and Cardiovascular Prevention, San Salvatore Hospital, 67100 L’Aquila, Italy
| |
Collapse
|
22
|
Nery Neto JADO, Yariwake VY, Câmara NOS, Andrade-Oliveira V. Enteroendocrine cells and gut hormones as potential targets in the crossroad of the gut-kidney axis communication. Front Pharmacol 2023; 14:1248757. [PMID: 37927592 PMCID: PMC10620747 DOI: 10.3389/fphar.2023.1248757] [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: 06/27/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023] Open
Abstract
Recent studies suggest that disruptions in intestinal homeostasis, such as changes in gut microbiota composition, infection, and inflammatory-related gut diseases, can be associated with kidney diseases. For instance, genomic investigations highlight how susceptibility genes linked to IgA nephropathy are also correlated with the risk of inflammatory bowel disease. Conversely, investigations demonstrate that the use of short-chain fatty acids, produced through fermentation by intestinal bacteria, protects kidney function in models of acute and chronic kidney diseases. Thus, the dialogue between the gut and kidney seems to be crucial in maintaining their proper function, although the factors governing this crosstalk are still emerging as the field evolves. In recent years, a series of studies have highlighted the significance of enteroendocrine cells (EECs) which are part of the secretory lineage of the gut epithelial cells, as important components in gut-kidney crosstalk. EECs are distributed throughout the epithelial layer and release more than 20 hormones in response to microenvironment stimuli. Interestingly, some of these hormones and/or their pathways such as Glucagon-Like Peptide 1 (GLP-1), GLP-2, gastrin, and somatostatin have been shown to exert renoprotective effects. Therefore, the present review explores the role of EECs and their hormones as regulators of gut-kidney crosstalk and their potential impact on kidney diseases. This comprehensive exploration underscores the substantial contribution of EEC hormones in mediating gut-kidney communication and their promising potential for the treatment of kidney diseases.
Collapse
Affiliation(s)
- José Arimatéa de Oliveira Nery Neto
- Bernardo’s Lab, Center for Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Victor Yuji Yariwake
- Bernardo’s Lab, Center for Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vinicius Andrade-Oliveira
- Bernardo’s Lab, Center for Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Mocanu A, Bogos RA, Lazaruc TI, Trandafir LM, Lupu VV, Ioniuc I, Alecsa M, Ivanov A, Lupu A, Starcea IM. Exploring a Complex Interplay: Kidney-Gut Axis in Pediatric Chronic Kidney Disease. Nutrients 2023; 15:3609. [PMID: 37630799 PMCID: PMC10457891 DOI: 10.3390/nu15163609] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The human intestinal microbiota is a highly intricate structure with a crucial role in promoting health and preventing disease. It consists of diverse microbial communities that inhabit the gut and contribute to essential functions such as food digestion, nutrient synthesis, and immune system development. The composition and function of the gut microbiota are influenced by a variety of factors, including diet, host genetics, and environmental features. In pediatric patients, the gut microbiota is particularly dynamic and vulnerable to disruption from endogenous and exogenous factors. Recent research has focused on understanding the interaction between the gut and kidneys. In individuals with chronic kidney disease, there is often a significant disturbance in the gut microbiota. This imbalance can be attributed to factors like increased levels of harmful toxins from the gut entering the bloodstream, inflammation, and oxidative stress. This review looks at what is known about the link between a child's gut-kidney axis, how dysbiosis, or an imbalance in the microbiome, affects chronic kidney disease, and what treatments, both pharmaceutical and non-pharmaceutical, are available for this condition.
Collapse
Affiliation(s)
- Adriana Mocanu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Nephrology Division, St. Mary’s Emergency Children Hospital, 700309 Iasi, Romania
| | - Roxana Alexandra Bogos
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Tudor Ilie Lazaruc
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Mihaela Trandafir
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Vasile Valeriu Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ileana Ioniuc
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mirabela Alecsa
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Anca Ivanov
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ancuta Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iuliana Magdalena Starcea
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Nephrology Division, St. Mary’s Emergency Children Hospital, 700309 Iasi, Romania
| |
Collapse
|
24
|
Hayeeawaema F, Muangnil P, Jiangsakul J, Tipbunjong C, Huipao N, Khuituan P. A novel model of adenine-induced chronic kidney disease-associated gastrointestinal dysfunction in mice: The gut-kidney axis. Saudi J Biol Sci 2023; 30:103660. [PMID: 37213695 PMCID: PMC10193294 DOI: 10.1016/j.sjbs.2023.103660] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/31/2023] [Accepted: 04/21/2023] [Indexed: 05/23/2023] Open
Abstract
Although constipation is a common complication of chronic kidney disease (CKD), there is no animal model that can be used to study the association between renal impairment and gastrointestinal function without interfering with the gastrointestinal tract of the model. Therefore, we determined whether adenine could induce CKD in association with gastrointestinal dysfunction. Six-week-old ICR mice were intraperitoneally injected with saline, 25, 50, or 75 mg adenine/kg body weight for 21 days. Blood urea nitrogen (BUN), plasma creatinine, and renal histopathology were evaluated. Defecation status was evaluated from defecation frequency and fecal water content. Colonic smooth muscle contraction was measured by the organ bath technique, and transepithelial electrical resistance (TEER) was measured using an Ussing chamber. In the 50 mg/kg treatment group, BUN and creatinine were significantly increased compared with control, and inflammatory cell infiltration, glomerular necrosis, tubular dilatation, and interstitial fibrosis were observed in renal tissues. Mice in this group also showed a significant decrease in defecation frequency, fecal water content, colonic motility index, and TEER. Overall, 50 mg/kg of adenine was the best dose to induce CKD with associated constipation and intestinal barrier impairment. Therefore, this adenine administration model can be recommended for CKD-associated gastrointestinal dysfunction research.
Collapse
Affiliation(s)
- Fittree Hayeeawaema
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla, Thailand
| | - Paradorn Muangnil
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla, Thailand
- Faculty of Veterinary Science, Prince of Songkla University, Thailand
| | | | - Chittipong Tipbunjong
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla, Thailand
| | - Nawiya Huipao
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla, Thailand
| | - Pissared Khuituan
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla, Thailand
- Corresponding author at: Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, 15 Karnjanavanich Rd., Hat Yai, Songkhla 90110, Thailand.
| |
Collapse
|
25
|
Hidayat AA, Gunawan VA, Iragama FR, Alfiansyah R, Hertanto DM, Tjempakasari A, Thaha M. Risk Factors and Clinical Characteristics of Acute Kidney Injury in Patients with COVID-19: A Systematic Review and Meta-Analysis. PATHOPHYSIOLOGY 2023; 30:233-247. [PMID: 37218918 DOI: 10.3390/pathophysiology30020020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
Acute kidney injury (AKI) is associated with a worse prognosis in coronavirus disease 2019 (COVID-19) patients. Identification of AKI, particularly in COVID-19 patients, is important for improving patients' management. The study aims to assess risk factors and comorbidities of AKI in COVID-19 patients. We systematically searched PubMed and DOAJ databases for relevant studies involving confirmed COVID-19 patients with data on risk factors and comorbidities of AKI. The risk factors and comorbidities were compared between AKI and non-AKI patients. A total of 30 studies involving 22385 confirmed COVID-19 patients were included. Male (OR: 1.74 (1.47, 2.05)), diabetes (OR: 1.65 (1.54, 1.76)), hypertension (OR: 1.82 (1.12, 2.95)), ischemic cardiac disease (OR: 1.70 (1.48, 1.95)), heart failure (OR: 2.29 (2.01, 2.59)), chronic kidney disease (CKD) (OR: 3.24 (2.20, 4.79)), chronic obstructive pulmonary disease (COPD) (OR: 1.86 (1.35, 2.57)), peripheral vascular disease (OR: 2.34 (1.20, 4.56)), and history of nonsteroidal anti-inflammatory drugs (NSAID) (OR: 1.59 (1.29, 1.98)) were independent risk factors associated with COVID-19 patients with AKI. Patients with AKI presented with proteinuria (OR: 3.31 (2.59, 4.23)), hematuria (OR: 3.25 (2.59, 4.08)), and invasive mechanical ventilation (OR: 13.88 (8.23, 23.40)). For COVID-19 patients, male gender, diabetes, hypertension, ischemic cardiac disease, heart failure, CKD, COPD, peripheral vascular disease, and history of use of NSAIDs are associated with a higher risk of AKI.
Collapse
Affiliation(s)
- Amal Arifi Hidayat
- Internal Medicine Resident, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Vania Azalia Gunawan
- Internal Medicine Resident, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Firda Rachmawati Iragama
- Internal Medicine Resident, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Rizky Alfiansyah
- Internal Medicine Resident, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Decsa Medika Hertanto
- Division of Nephrology-Hypertension, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Artaria Tjempakasari
- Division of Nephrology-Hypertension, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Mochammad Thaha
- Division of Nephrology-Hypertension, Department of Internal Medicine, Dr. Soetomo Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
26
|
Temel HY, Kaymak Ö, Kaplan S, Bahcivanci B, Gkoutos GV, Acharjee A. Role of microbiota and microbiota-derived short-chain fatty acids in PDAC. Cancer Med 2023; 12:5661-5675. [PMID: 36205023 PMCID: PMC10028056 DOI: 10.1002/cam4.5323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive lethal diseases among other cancer types. Gut microbiome and its metabolic regulation play a crucial role in PDAC. Metabolic regulation in the gut is a complex process that involves microbiome and microbiome-derived short-chain fatty acids (SCFAs). SCFAs regulate inflammation, as well as lipid and glucose metabolism, through different pathways. This review aims to summarize recent developments in PDAC in the context of gut and oral microbiota and their associations with short-chain fatty acid (SCFA). In addition to this, we discuss possible therapeutic applications using microbiota in PDAC.
Collapse
Affiliation(s)
- Hülya Yılmaz Temel
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Öznur Kaymak
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Seren Kaplan
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Basak Bahcivanci
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| |
Collapse
|
27
|
Tain YL, Hsu CN. Role of the Gut Microbiota in Children with Kidney Disease. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020269. [PMID: 36832398 PMCID: PMC9955067 DOI: 10.3390/children10020269] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
Disruption of the composition and structure of the gut microbiota, namely dysbiosis, dictates the pathophysiology of kidney diseases. The bidirectional kidney-gut axis is of interest in chronic kidney disease (CKD); the uremic milieu leads to intestinal dysbiosis and gut microbial metabolites and toxins implicated in the loss of kidney function and increased comorbidity burden. Considering that kidney diseases can originate in childhood or even earlier in fetal life, identification of the pathogenetic connection between gut microbiota dysbiosis and the development of pediatric renal diseases deserves more attention. This review concentrates on the pathogenic link between dysbiotic gut microbiota and pediatric renal diseases, covering CKD, kidney transplantation, hemodialysis and peritoneal dialysis, and idiopathic nephrotic syndrome. Gut microbiota-targeted therapies including dietary intervention, probiotics, prebiotics, postbiotics and fecal microbial transplantation are discussed for their potential for the treatment of pediatric renal diseases. A deeper understanding of gut microbiota in pediatric renal diseases will aid in developing innovative gut microbiota-targeted interventions for preventing or attenuating the global burden of kidney diseases.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-975-368-975; Fax: +886-7733-8009
| |
Collapse
|
28
|
Liu W, Huang J, Liu T, Hu Y, Shi K, Zhou Y, Zhang N. Changes in gut microbial community upon chronic kidney disease. PLoS One 2023; 18:e0283389. [PMID: 36952529 PMCID: PMC10035866 DOI: 10.1371/journal.pone.0283389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/08/2023] [Indexed: 03/25/2023] Open
Abstract
With the increasing incidence and mortality of chronic kidney disease (CKD), targeted therapies for CKD have been explored constantly. The important role of gut microbiota on CKD has been emphasized increasingly, it is necessary to analyze the metabolic mechanism of CKD patients from the perspective of gut microbiota. In this study, bioinformatics was used to analyze the changes of gut microbiota between CKD and healthy control (HC) groups using 315 samples from NCBI database. Diversity analysis showed significant changes in evenness compared to the HC group. PCoA analysis revealed significant differences between the two groups at phylum level. In addition, the F/B ratio was higher in CKD group than in HC group, suggesting the disorder of gut microbiota, imbalance of energy absorption and the occurrence of metabolic syndrome in CKD group. The study found that compared with HC group, the abundance of bacteria associated with impaired kidney was increased in CKD group, such as Ralstonia and Porphyromonas, which were negatively associated with eGFR. PICRUSt2 was used to predict related functions and found that different pathways between the two groups were mainly related to metabolism, involving the metabolism of exogenous and endogenous substances, as well as Glycerophospholipid metabolism, which provided evidence for exploring the relationship between gut microbiota and lipid metabolism. Therefore, in subsequent studies, special attention should be paid to these bacteria and metabolic pathway, and animal experiments and metabolomics studies should be conducted explore the association between bacterial community and CKD, as well as the therapeutic effects of these microbial populations on CKD.
Collapse
Affiliation(s)
- Wu Liu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaqi Huang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Tong Liu
- School of Water Resources and Environment, China University of Geosciences (Beijing), Beijing, China
| | - Yutian Hu
- School of Water Resources and Environment, China University of Geosciences (Beijing), Beijing, China
| | - Kaifeng Shi
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Zhou
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate Student, Beijing University of Chinese Medicine, Beijing, China
| | - Ning Zhang
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Probiotic Bifidobacteria Mitigate the Deleterious Effects of para-Cresol in a Drosophila melanogaster Toxicity Model. mSphere 2022; 7:e0044622. [PMID: 36321825 PMCID: PMC9769938 DOI: 10.1128/msphere.00446-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Renal impairment associated with chronic kidney disease (CKD) causes the buildup of uremic toxins that are deleterious to patient health. Current therapies that manage toxin accumulation in CKD offer an incomplete therapeutic effect against toxins such as para-cresol (p-cresol) and p-cresyl sulfate. Probiotic therapies can exploit the wealth of microbial diversity to reduce toxin accumulation. Using in vitro culture techniques, strains of lactobacilli and bifidobacteria from a 24-strain synbiotic were investigated for their ability to remove p-cresol. Four strains of bifidobacteria internalized p-cresol from the extracellular environment. The oral supplementation of these toxin-clearing probiotics was more protective than control strains in a Drosophila melanogaster toxicity model. Bifidobacterial supplementation was also associated with higher abundance of lactobacilli in the gut microbiota of p-cresol-exposed flies. The present findings suggest that these strains might reduce p-cresol in the gut in addition to increasing the prevalence of other beneficial bacteria, such as lactobacilli, and should be tested clinically to normalize the dysbiotic gut microbiota observed in CKD patients. IMPORTANCE Chronic kidney disease (CKD) affects approximately 10% of the global population and has limited treatment options. The accumulation of gut microbiota-derived uremic toxins, such as para-cresol (p-cresol) and p-cresyl sulfate, is associated with the onset of comorbidities (i.e., atherosclerosis and cognitive disorders) in CKD. Unfortunately, dialysis, the gold standard therapy is unable to remove these toxins from the bloodstream due to their highly protein-bound nature. Some strains of Bifidobacterium have metabolic properties that may be useful in managing uremic toxicity. Using a Drosophila model, the present work highlights why dosing with certain probiotic strains may be clinically useful in CKD management.
Collapse
|
30
|
Traditional Chinese Medicine: An Exogenous Regulator of Crosstalk between the Gut Microbial Ecosystem and CKD. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7940684. [DOI: 10.1155/2022/7940684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022]
Abstract
Chronic kidney disease (CKD) is often accompanied by an imbalance in the gut microbial ecosystem. Notably, the imbalanced gut microbiota and impaired intestinal barrier are the keys to the crosstalk between the gut microbial ecosystem and CKD, which was the central point of previous studies. Traditional Chinese medicine (TCM) has shown considerable efficacy in the treatment of CKD. However, the therapeutic mechanisms have not been fully elucidated. In this review, we explored therapeutic mechanisms by which TCM improved CKD via the gut microbial ecosystem. In particular, we focused on the restored gut microbiota (i.e., short-chain fatty acid- and uremic toxin-producing bacteria), improved gut-derived metabolites (i.e., short-chain fatty acid, indoxyl sulfate, p-Cresyl sulfate, and trimethylamine-N-oxide), and intestinal barrier (i.e., permeability and microbial translocation) as therapeutic mechanisms. The results found that the metabolic pattern of gut microbiota and the intestinal barrier were improved through TCM treatment. Moreover, the microbiota-transfer study confirmed that part of the protective effect of TCM was dependent on gut microbiota, especially SCFA-producing bacteria. In conclusion, TCM may be an important exogenous regulator of crosstalk between the gut microbial ecosystem and CKD, which was partly attributable to the mediation of microbiota-targeted intervention.
Collapse
|
31
|
da Silva TCA, dos Santos Gonçalves JA, Souza LACE, Lima AA, Guerra-Sá R. The correlation of the fecal microbiome with the biochemical profile during menopause: a Brazilian cohort study. BMC Womens Health 2022; 22:499. [PMID: 36474222 PMCID: PMC9724392 DOI: 10.1186/s12905-022-02063-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hormonal, biochemical, and metabolic changes after menopause may alter the quality of life of women, leading to vasomotor, psychological, and genitourinary symptoms, and changes in their gut microbiota, which regulates estrogen levels through the estroboloma. Fecal samples were used to investigate the changes in the gut microbiota during aging and hormonal changes in women. A balanced gut microbiota has been associated with health or disease conditions and remains poorly understood after menopause. This study identified the fecal microbiota, and their association with biochemical and hormonal parameters of a cohort of women in the climacteric in the city of Ouro Preto-MG, Brazil. METHODS A total of 102 women aged 40 to 65 years old were recruited and distributed into three groups according to the STRAW criteria for reproductive stage: reproductive (n = 18), premenopausal (n = 26), and postmenopausal (n = 58). Blood samples were collected to measure their serum biochemical and hormone levels, and the participants answered a questionnaire. The gut microbiota was analyzed from fecal samples by qPCR using the genera Bifidobacterium, Bacteroides, Lactobacillus, and Clostridium. RESULTS The following parameters showed differences among the groups: total cholesterol, triglycerides, VLDL, ApoB, urea, calcium, uric acid, and alkaline phosphatase (p < 0.05). qPCR revealed the genus Clostridium to be the most abundant in all three groups. In the reproductive age group, the significant correlations were: Bacteroides with glucose (r = -0.573 p = 0.0129), and SDHEA (r = -0.583 p = 0.0111). For the premenopausal group, they were: Bifidobacteria with total cholesterol (r = 0.396 p = 0.0451), LDL (r = 0.393 p = 0.0468), ApoB (r = 0.411 p = 0.0368); Lactobacillus and calcium (r = 0.443 p = 0.0232), ALP (r = 0.543 p = 0.0041), LPa (r =-0.442 p = 0.02336); and Bacteroides and urea (r =-0.461 p = 0.0176). In the postmenopausal group, they were Bifidobacterium and ALP (r =-0.315 p = 0.0159), Lactobacillus and urea (r =-0.276 p = 0.0356), and Clostridium and beta estradiol (r =-0.355 p = 0.0062). CONCLUSION In conclusion, the hormonal and metabolic changes during menopause in the population studied were accompanied by a significant change in the fecal microbiota, especially of the genus Clostridium.
Collapse
Affiliation(s)
- Thayane Christine Alves da Silva
- grid.411213.40000 0004 0488 4317Graduate Program in Biological Sciences - Biological Sciences Research Center, Federal University of Ouro Preto, Morro Do Cruzeiro, Ouro Preto, Minas Gerais Brazil ,grid.411213.40000 0004 0488 4317Laboratory of Biochemistry and Molecular Biology (LBBM), Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Minas Gerais Brazil
| | - Jennefer Aparecida dos Santos Gonçalves
- grid.411213.40000 0004 0488 4317Laboratory of Biochemistry and Molecular Biology (LBBM), Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Minas Gerais Brazil
| | - Laura Alves Cota e Souza
- grid.411213.40000 0004 0488 4317Graduate Program in Pharmaceutical Sciences (CiPharma), School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Minas Gerais Brazil
| | - Angélica Alves Lima
- grid.411213.40000 0004 0488 4317Graduate Program in Pharmaceutical Sciences (CiPharma), School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Minas Gerais Brazil
| | - R. Guerra-Sá
- grid.411213.40000 0004 0488 4317Graduate Program in Biological Sciences - Biological Sciences Research Center, Federal University of Ouro Preto, Morro Do Cruzeiro, Ouro Preto, Minas Gerais Brazil ,grid.411213.40000 0004 0488 4317Laboratory of Biochemistry and Molecular Biology (LBBM), Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Minas Gerais Brazil
| |
Collapse
|
32
|
Paul P, Kaul R, Chaari A. Renal Health Improvement in Diabetes through Microbiome Modulation of the Gut-Kidney Axis with Biotics: A Systematic and Narrative Review of Randomized Controlled Trials. Int J Mol Sci 2022; 23:14838. [PMID: 36499168 PMCID: PMC9740604 DOI: 10.3390/ijms232314838] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/13/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Diabetes mellitus is the most common endocrine disorder worldwide, with over 20% of patients ultimately developing diabetic kidney disease (DKD), a complex nephropathic complication that is a leading cause of end-stage renal disease. Various clinical trials have utilized probiotics, prebiotics, and synbiotics to attempt to positively modulate the gut microbiome via the gut-kidney axis, but consensus is limited. We conducted a multi-database systematic review to investigate the effect of probiotics, prebiotics, and synbiotics on various biomarkers of renal health in diabetes, based on studies published through 10 April 2022. Adhering to the Cochrane Collaboration and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, relevant articles were systematically screened and extracted by independent reviewers; subsequently, results were systematically compiled, analyzed, and expanded through a narrative discussion. A total of 16 publications encompassing 903 diabetic individuals met the inclusion criteria. Our findings show that some studies report statistically significant changes in common renal markers, such as serum creatinine, estimated glomerular filtration rate, blood urea nitrogen/urea, microalbuminuria, and uric acid, but not on serum albumin, sodium, potassium, phosphorous, or total urine protein. Interestingly, these nutraceuticals seem to increase serum uric acid concentrations, an inflammatory marker usually associated with decreased renal health. We found that probiotics from the Lactobacillus and Bifidobacterium families were the most investigated, followed by Streptococcus thermophilus. Prebiotics including inulin, galacto-oligosaccharide, and resistant dextrin were also examined. The single-species probiotic soymilk formulation of Lactobacillus plantarum A7 possessed effects on multiple renal biomarkers in DKD patients without adverse events. We further investigated the optimum nutraceutical formulation, discussed findings from prior studies, described the gut-kidney axis in diabetes and DKD, and finally commented on some possible mechanisms of action of these nutraceuticals on renal health in diabetics. Although probiotics, prebiotics, and synbiotics have shown some potential in ameliorating renal health degradation in diabetes via gut-kidney axis crosstalk, larger and more convincing trials with focused objectives and next-generation nutraceutical formulations are required to investigate their possible role as adjunct therapy in such patients.
Collapse
Affiliation(s)
- Pradipta Paul
- Medical Education Division, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation—Education City, Doha P.O. Box 24144, Qatar
| | - Ridhima Kaul
- Medical Education Division, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation—Education City, Doha P.O. Box 24144, Qatar
| | - Ali Chaari
- Premedical Division, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation—Education City, Doha P.O. Box 24144, Qatar
| |
Collapse
|
33
|
Kuznetzova AB, Prazdnova EV, Chistyakov VA, Kutsevalova OY, Batiushin MM. Are Probiotics Needed in Nephrology? NEPHROLOGY (SAINT-PETERSBURG) 2022; 26:18-30. [DOI: 10.36485/1561-6274-2022-26-4-18-30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- A. B. Kuznetzova
- Academy of Biology and Biotechnology named after D.I. Ivanovsky, Southern Federal University
| | - E. V. Prazdnova
- Academy of Biology and Biotechnology named after D.I. Ivanovsky, Southern Federal University
| | - V. A. Chistyakov
- Academy of Biology and Biotechnology named after D.I. Ivanovsky, Southern Federal University
| | - O. Yu. Kutsevalova
- Federal State Budgetary Institution "National Medical Research Center of Oncology"
| | | |
Collapse
|
34
|
Xu J, Moore BN, Pluznick JL. Short-Chain Fatty Acid Receptors and Blood Pressure Regulation: Council on Hypertension Mid-Career Award for Research Excellence 2021. Hypertension 2022; 79:2127-2137. [PMID: 35912645 PMCID: PMC9458621 DOI: 10.1161/hypertensionaha.122.18558] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gut microbiome influences host physiology and pathophysiology through several pathways, one of which is microbial production of chemical metabolites which interact with host signaling pathways. Short-chain fatty acids (SCFAs) are a class of gut microbial metabolites known to activate multiple signaling pathways in the host. Growing evidence indicates that the gut microbiome is linked to blood pressure, that SCFAs modulate blood pressure regulation, and that delivery of exogenous SCFAs lowers blood pressure. Given that hypertension is a key risk factor for cardiovascular disease, the examination of novel contributors to blood pressure regulation has the potential to lead to novel approaches or treatments. Thus, this review will discuss SCFAs with a focus on their host G protein-coupled receptors including GPR41 (G protein-coupled receptor 41), GPR43, and GPR109A, as well as OLFR78 (olfactory receptor 78) and OLFR558. This includes a discussion of the ligand profiles, G protein coupling, and tissue distribution of each receptor. We will also review phenotypes relevant to blood pressure regulation which have been reported to date for Gpr41, Gpr43, Gpr109a, and Olfr78 knockout mice. In addition, we will consider how SCFA signaling influences physiology at baseline, and, how SCFA signaling may contribute to blood pressure regulation in settings of hypertension. In sum, this review will integrate current knowledge regarding how SCFAs and their receptors regulate blood pressure.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Brittni N. Moore
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jennifer L. Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
35
|
Bhargava S, Merckelbach E, Noels H, Vohra A, Jankowski J. Homeostasis in the Gut Microbiota in Chronic Kidney Disease. Toxins (Basel) 2022; 14:648. [PMID: 36287917 PMCID: PMC9610479 DOI: 10.3390/toxins14100648] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota consists of trillions of microorganisms, fulfilling important roles in metabolism, nutritional intake, physiology and maturation of the immune system, but also aiding and abetting the progression of chronic kidney disease (CKD). The human gut microbiome consists of bacterial species from five major bacterial phyla, namely Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. Alterations in the members of these phyla alter the total gut microbiota, with a decline in the number of symbiotic flora and an increase in the pathogenic bacteria, causing or aggravating CKD. In addition, CKD-associated alteration of this intestinal microbiome results in metabolic changes and the accumulation of amines, indoles and phenols, among other uremic metabolites, which have a feedforward adverse effect on CKD patients, inhibiting renal functions and increasing comorbidities such as atherosclerosis and cardiovascular diseases (CVD). A classification of uremic toxins according to the degree of known toxicity based on the experimental evidence of their toxicity (number of systems affected) and overall experimental and clinical evidence was selected to identify the representative uremic toxins from small water-soluble compounds, protein-bound compounds and middle molecules and their relation to the gut microbiota was summarized. Gut-derived uremic metabolites accumulating in CKD patients further exhibit cell-damaging properties, damage the intestinal epithelial cell wall, increase gut permeability and lead to the translocation of bacteria and endotoxins from the gut into the circulatory system. Elevated levels of endotoxins lead to endotoxemia and inflammation, further accelerating CKD progression. In recent years, the role of the gut microbiome in CKD pathophysiology has emerged as an important aspect of corrective treatment; however, the mechanisms by which the gut microbiota contributes to CKD progression are still not completely understood. Therefore, this review summarizes the current state of research regarding CKD and the gut microbiota, alterations in the microbiome, uremic toxin production, and gut epithelial barrier degradation.
Collapse
Affiliation(s)
- Shruti Bhargava
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, 52062 Aachen, Germany
| | - Erik Merckelbach
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, 52062 Aachen, Germany
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, 52062 Aachen, Germany
| | - Ashima Vohra
- Institute of Home Economics, Delhi University, Delhi 110021, India
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, 52062 Aachen, Germany
- Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, 6211 Maastricht, The Netherlands
| |
Collapse
|
36
|
Lin Y, Ma B, Yang Y, Chen Y, Huang J, Li W, Yu X, Liang L. Impaired kidney function biomarkers and risk of severe COVID-19: Analysis of population-based cohort data. Mol Genet Genomic Med 2022; 10:e2047. [PMID: 36124564 PMCID: PMC9538291 DOI: 10.1002/mgg3.2047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with impaired kidney function were found at a high risk of COVID-19 hospitalization and mortality in many observational, cross-sectional, and hospital-based studies, but evidence from large-scale prospective cohorts has been lacking. We aimed to examine the association of kidney function-related biomarkers and their genetic predisposition with the risk of developing severe COVID-19 in population-based data. METHODS We analyzed data from UK Biobank to examine the prospective association of abnormal kidney function biomarkers with severe COVID-19, defined by laboratory-confirmed COVID-19 hospitalizations. Using genotype data, we constructed polygenic risk scores (PRS) to represent an individual's overall genetic risk for these biomarkers. We also identified tipping points where the risk of severe COVID-19 began to increase significantly for each biomarker. RESULTS Of the 502,506 adults, 1650 (0.32%) were identified as severe COVID-19, before August 12, 2020. High levels of cystatin C (OR: 1.3; 95% CI: 1.2-1.5; FDR = 1.5 × 10-5 ), serum creatinine (OR: 1.7; 95% CI: 1.3-2.1; p = 3.5 × 10-4 ; FDR = 3.5 × 10-4 ), microalbuminuria (OR: 1.4; 95% CI: 1.2-1.6; FDR = 4 × 10-4 ), and UACR (urinary albumin creatinine ratio; OR: 1.4; 95% CI: 1.2-1.6; p = 3.5 × 10-4 ; FDR = 3.5 × 10-4 ) were found significantly associated with severe COVID-19. Individuals with top 10% of PRS for elevated cystatin C, urate, and microalbuminuria had 28% to 43% higher risks of severe COVID-19 than individuals with bottom 30% PRS (p < 0.05). Tipping-point analyses further supported that severe COVID-19 could occur even when the values of cystatin C, urate (male), and microalbuminuria were within their normal value ranges (OR >1.1, p < 0.05). CONCLUSIONS Findings from this study might point to new directions for clinicians and policymakers in optimizing risk-stratification among patients based on polygenic risk estimation and tipping points of kidney function markers. Our results call for further investigation to develop a better strategy to prevent severe COVID-19 outcomes among patients with genetic predisposition to impaired kidney function. These findings could provide a new tool for clinicians and policymakers in the future especially if we need to live with COVID-19 for a long time.
Collapse
Affiliation(s)
- Yifei Lin
- West China HospitalSichuan UniversityChengduChina,Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Baoshan Ma
- College of Information Science and TechnologyDalian Maritime UniversityDalianChina
| | - Yingxi Yang
- Department of StatisticsSun Yat‐sen UniversityGuangzhouChina
| | - Yuxiang Chen
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Jin Huang
- West China HospitalSichuan UniversityChengduChina
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine/Institute of Respiratory Health, Frontiers Science Center for Disease‐related Molecular Network/Precision Medicine Research Center/The Research Units of West China, Chinese Academy of Medical Sciences, West China HospitalSichuan UniversityChengduChina
| | - Xueqing Yu
- Department of NephrologyGuangdong Provincial People's HospitalGuangzhouChina
| | - Liming Liang
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA,Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
37
|
Tan J, Chen M, Wang Y, Tang Y, Qin W. Emerging trends and focus for the link between the gastrointestinal microbiome and kidney disease. Front Cell Infect Microbiol 2022; 12:946138. [PMID: 36046740 PMCID: PMC9420905 DOI: 10.3389/fcimb.2022.946138] [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: 05/17/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
The clinical value of the relationship between gastrointestinal microbiome imbalance and its corresponding interventions with kidney disease is emerging. This study describes the hotspots and evolution of gastrointestinal microbiome and kidney disease research over the past three decades by scientific collaboration networks and finally predicts future trends in the field through bibliometric analysis and visualization studies. CiteSpace was used to explore the original articles from January 1990 to August 2021 to visualize the knowledge network of journals, countries, institutions, authors, references, and keywords in this field. Publications were extracted from Web of Science Core Collection database using the terms “gastrointestinal microbiome” and “kidney disease” (and their synonyms in MeSH). A total of 2145 publications with 93880 references in 102 journals were included in the analyses. The number of studies combining gastrointestinal microbiomes with kidney diseases has increased significantly over the past two decades. The United States is the leading country in the number of documents, and the leading institution is the Cleveland Clinic. The most landmark articles in the field are on chronic renal failure, L-Carnitin, and cardiovascular disease. The pathogenesis of uremia toxin is an emerging trend in gastrointestinal microbiomes and kidney diseases. In addition, probiotic or synbiotic supplements have strong clinical value in adjusting abnormal intestinal symbiotic environments. This study demonstrates a growing understanding of the interaction between gut microbiota and kidney disease over time. Using microbial supplements to improve the living conditions of kidney disease patients is a promising and hot research focus. Based on publications extracted from the database, this study may provide clinicians and researchers with valuable information to identify potential collaborators and partner institutions and better predict their dynamic progression.
Collapse
Affiliation(s)
- Jiaxing Tan
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Ming Chen
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yutong Wang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yi Tang
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
- *Correspondence: Yi Tang, ; Wei Qin,
| | - Wei Qin
- Division of Nephrology, Department of Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
- *Correspondence: Yi Tang, ; Wei Qin,
| |
Collapse
|
38
|
Rahman MM, Islam F, -Or-Rashid MH, Mamun AA, Rahaman MS, Islam MM, Meem AFK, Sutradhar PR, Mitra S, Mimi AA, Emran TB, Fatimawali, Idroes R, Tallei TE, Ahmed M, Cavalu S. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front Cell Infect Microbiol 2022; 12:903570. [PMID: 35795187 PMCID: PMC9251340 DOI: 10.3389/fcimb.2022.903570] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
In the last two decades, considerable interest has been shown in understanding the development of the gut microbiota and its internal and external effects on the intestine, as well as the risk factors for cardiovascular diseases (CVDs) such as metabolic syndrome. The intestinal microbiota plays a pivotal role in human health and disease. Recent studies revealed that the gut microbiota can affect the host body. CVDs are a leading cause of morbidity and mortality, and patients favor death over chronic kidney disease. For the function of gut microbiota in the host, molecules have to penetrate the intestinal epithelium or the surface cells of the host. Gut microbiota can utilize trimethylamine, N-oxide, short-chain fatty acids, and primary and secondary bile acid pathways. By affecting these living cells, the gut microbiota can cause heart failure, atherosclerosis, hypertension, myocardial fibrosis, myocardial infarction, and coronary artery disease. Previous studies of the gut microbiota and its relation to stroke pathogenesis and its consequences can provide new therapeutic prospects. This review highlights the interplay between the microbiota and its metabolites and addresses related interventions for the treatment of CVDs.
Collapse
|
39
|
Chiang WF, Hsiao PJ, Chan JS. Vitamin D for Recovery of COVID-19 in Patients With Chronic Kidney Disease. Front Nutr 2022; 9:930176. [PMID: 35782942 PMCID: PMC9240470 DOI: 10.3389/fnut.2022.930176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/26/2022] [Indexed: 12/22/2022] Open
Abstract
The severity of coronavirus disease 2019 (COVID-19) is determined not only by viral damage to cells but also by the immune reaction in the host. In addition to therapeutic interventions that target the viral infection, immunoregulation may be helpful in the management of COVID-19. Vitamin D exerts effects on both innate and adaptive immunity and subsequently modulates immune responses to bacteria and viruses. Patients with chronic kidney disease (CKD) frequently have vitamin D deficiency and increased susceptibility to infection, suggesting a potential role of vitamin D in this vulnerable population. In this paper, we review the alterations of the immune system, the risk of COVID-19 infections and mechanisms of vitamin D action in the pathogenesis of COVID-19 in CKD patients. Previous studies have shown that vitamin D deficiency can affect the outcomes of COVID-19. Supplementing vitamin D during treatment may be protective against COVID-19. Future studies, including randomized control trials, are warranted to determine the effect of vitamin D supplementation on the recovery from COVID-19 in CKD patients.
Collapse
Affiliation(s)
- Wen-Fang Chiang
- Division of Nephrology, Department of Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Medicine, Armed Forces Taoyuan General Hospital, Taoyuan, Taiwan
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
40
|
Dai Y, Quan J, Xiong L, Luo Y, Yi B. Probiotics improve renal function, glucose, lipids, inflammation and oxidative stress in diabetic kidney disease: a systematic review and meta-analysis. Ren Fail 2022; 44:862-880. [PMID: 35611435 PMCID: PMC9154786 DOI: 10.1080/0886022x.2022.2079522] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AIMS The role of probiotics in the management of diabetic kidney disease (DKD) has been shown. Several current trials are investigating the effect of probiotics, which are widely used to modulate biomarkers of renal function, glucose, lipids, inflammation and oxidative stress in patients with DKD. However, their findings are controversial. This study aimed to systematically evaluate the impact of probiotics on patients with DKD via meta-analysis. METHODS PubMed, The Cochrane Library, Web of Science, Scopus, Embase, China National Knowledge Infrastructure, Chinese Wanfang Database and Chinese VIP Database were searched for relevant studies from the establishment of these databases to September 2021. The pooled results evaluated the impact of probiotics on renal function, glucose, lipids, inflammation and oxidative stress indicators in patients with DKD. Additionally, subgroup analysis was performed based on intervention duration, probiotic dose and probiotic consumption patterns, respectively. RESULTS Ten trials that included 552 participants were identified for analysis. Compared with the controls, probiotics significantly decreased serum creatinine (Scr) [WMD = -0.17 mg/dL; 95%CI = -0.29, -0.05; p = 0.004], blood urea nitrogen (BUN) [WMD = -1.36 mg/dL; 95%CI = -2.20, -0.52; p = 0.001], cystatin C (Cys C) [WMD = -29.50 ng/mL; 95%CI = -32.82, -26.18; p < 0.00001], urinary albumin/creatinine ratio (UACR) [WMD = -16.05 mg/g; 95%CI = -27.12, -4.99; p = 0.004] and natrium (Na) [WMD = -0.94 mmol/L; 95%CI = -1.82, -0.05; p = 0.04] in patients with DKD. Enhanced glycemic control was observed in patients with DKD receiving probiotics compared with controls, as demonstrated by reduced levels of fasting plasma glucose (FPG), hemoglobin A1c (HbA1c), homeostasis model of assessment-estimated insulin resistance (HOMA-IR), and increased quantitative insulin sensitivity check index (QUICKI). Probiotics affected lipid metabolism parameters with decreasing triglycerides (TG), total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-c) levels in patients with DKD. Probiotics could also could improve inflammation and oxidative stress by decreasing high-sensitivity C-reactive protein (hs-CRP), plasma malondialdehyde (MDA), total antioxidant capacity (TAC), glutathione (GSH) and nitric oxide (NO). Additionally, subgroup analysis showed that those who received multiple species probiotics had a statistically significant difference in BUN, FPG, HOMA-IR, high-density lipoprotein cholesterol (HDL-c), MDA, TAC, and NO. Meanwhile, Scr, LDL-c, HDL-c, MDA, and TAC were ameliorated when the intervention duration was more than eight weeks and BUN, FPG, HOMA-IR, and MDA were improved when the probiotic dose was greater than four billion CFU/day. CONCLUSIONS Our analysis revealed that probiotics could delay the progression of renal function injury, improve glucose and lipid metabolism, and reduce inflammation and oxidative stress in patients with DKD. Subgroup analysis showed that intervention duration, probiotic dose and probiotic consumption patterns had an effect of probiotics on outcomes.
Collapse
Affiliation(s)
- Yali Dai
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, China.,Department of Nephrology, Yueyang People's Hospital, Yueyang Hospital affiliated to Hunan Normal University, Yueyang, China
| | - Jingjing Quan
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lianlian Xiong
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yanfang Luo
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Bin Yi
- Department of Nephrology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Parsegian K, Randall D, Curtis M, Ioannidou E. Association between periodontitis and chronic kidney disease. Periodontol 2000 2022; 89:114-124. [PMID: 35244955 DOI: 10.1111/prd.12431] [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] [Indexed: 12/21/2022]
Abstract
Periodontitis and chronic kidney disease are chronic conditions with high community prevalence across the world. Patients with chronic kidney disease have been noted to have a high burden of periodontitis, and several shared risk factors have been associated with the prevalence and severity of both conditions. However, the precise relationship between the two conditions, and the extent to which each may contribute to the development of the other, remains a matter of debate. The goals of the present work were to: (a) provide the most current and relevant literature overview of the association between periodontitis and chronic kidney disease; (b) explore mechanisms underlying this association; and (c) determine if evidence exists for an independent association between these conditions. We also assessed whether improved oral hygiene and periodontal treatment could reduce the risk of developing chronic kidney disease and, if so, what protocols these strategies involve. Finally, we aimed to reveal gaps in our current knowledge to delineate the directions of future research. Although the exact relationship between these two conditions has not yet been defined, we highlight the importance of the interprofessional interaction between dental practitioners and the nephrology team and the importance of oral health assessment in the management of chronic kidney disease.
Collapse
Affiliation(s)
- Karo Parsegian
- Department of Surgical Dentistry, Division of Periodontics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David Randall
- William Harvey Research Institute, Charterhouse Square Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mike Curtis
- Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Hospital, London, UK
| | - Effie Ioannidou
- Department of Oral Health and Diagnostic Sciences, Division of Periodontology, Dental Clinical Research Center, School of Dental Medicine, University of Connecticut Health, Farmington, Connecticut, USA
| |
Collapse
|
42
|
Zsom L, Zsom M, Salim SA, Fülöp T. Estimated Glomerular Filtration Rate in Chronic Kidney Disease: A Critical Review of Estimate-Based Predictions of Individual Outcomes in Kidney Disease. Toxins (Basel) 2022; 14:127. [PMID: 35202154 PMCID: PMC8875627 DOI: 10.3390/toxins14020127] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is generally regarded as a final common pathway of several renal diseases, often leading to end-stage kidney disease (ESKD) and a need for renal replacement therapy. Estimated GFR (eGFR) has been used to predict this outcome recognizing its robust association with renal disease progression and the eventual need for dialysis in large, mainly cross-sectional epidemiological studies. However, GFR is implicitly limited as follows: (1) GFR reflects only one of the many physiological functions of the kidney; (2) it is dependent on several non-renal factors; (3) it has intrinsic variability that is a function of dietary intake, fluid and cardiovascular status, and blood pressure especially with impaired autoregulation or medication use; (4) it has been shown to change with age with a unique non-linear pattern; and (5) eGFR may not correlate with GFR in certain conditions and disease states. Yet, many clinicians, especially our non-nephrologist colleagues, tend to regard eGFR obtained from a simple laboratory test as both a valid reflection of renal function and a reliable diagnostic tool in establishing the diagnosis of CKD. What is the validity of these beliefs? This review will critically reassess the limitations of such single-focused attention, with a particular focus on inter-individual variability. What does science actually tell us about the usefulness of eGFR in diagnosing CKD?
Collapse
Affiliation(s)
- Lajos Zsom
- Fresenius Medical Care, Cegléd Dialysis Center, Törteli u 1-3, 2700 Cegléd, Hungary
| | - Marianna Zsom
- Department of Medicine, St. Rókus Hospital, Rókus u 10, 6500 Baja, Hungary;
| | - Sohail Abdul Salim
- Department of Medicine, Division of Nephrology, University of Mississippi, 2500 N State St., Jackson, MS 39216, USA;
| | - Tibor Fülöp
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, 96 Jonathan Lucas Street, MSC 629, CSB 822, Charleston, SC 29425, USA
- Medicine Services, Ralph H. Johnson VA Medical Center, 109 Bee St., Charleston, SC 29401, USA
| |
Collapse
|
43
|
Li J, Li H, Deng W, Meng L, Gong W, Yao H. The Effect of Combination Use of Hemodialysis and Hemoperfusion on Microinflammation in Elderly Patients with Maintenance Hemodialysis. Blood Purif 2022; 51:739-746. [DOI: 10.1159/000518857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/24/2021] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Patients with maintenance hemodialysis (MHD) generally have a microinflammatory state. The aim of this study was to investigate the effects of hemodialysis (HD) combined with hemoperfusion (HP) on microinflammatory state in elderly patients with MHD. <b><i>Methods:</i></b> One hundred and fifty elderly patients with MHD were randomly divided into the control group and the observation group. The control group received simple HD treatment, and the observation group received combined HD + HP treatment on the basis of the control group. After 6 months of continuous treatment, the patients were evaluated to compare the quality of life, inflammation, adverse reactions, and nutritional indicators in the 2 groups before and after treatment. <b><i>Results:</i></b> There was no significant difference in the quality of life between the 2 groups before treatment. After treatment, the scores of psychological aspects, physiological aspects, social aspects, environmental aspects, and independent ability in the observation group were higher than those in the control group, with statistical significance (<i>p</i> < 0.05). There was no statistical significance in the level of inflammation between 2 groups before treatment. After treatment, the levels of hs-CRP, Hcy, IL-6, and TNF-α in the observation group were significantly lower than those in the control group, with statistical significance (<i>p</i> < 0.05). The incidence of dry mouth, skin reaction, neuritis, and subcutaneous tissue fibrosis in the observation group was lower than that in the control group, with statistical significance (<i>p</i> < 0.05). There was no statistical significance in nutritional level indexes between 2 groups before treatment (<i>p</i><sub>1</sub> > 0.05). After treatment, the levels of hemoglobin, total protein, albumin, and transferrin in the observation group were significantly higher than those in the control group, with statistical significance (<i>p</i> < 0.05). <b><i>Conclusion:</i></b> The clinical effect of HD combined with HP in elderly MHD patients is significant, which can effectively reduce the incidence of adverse reactions and inflammation in the patients and improve the quality of life and nutritional indicators of the patients.
Collapse
|
44
|
Yan Z, Wang G, Shi X. Advances in the Progression and Prognosis Biomarkers of Chronic Kidney Disease. Front Pharmacol 2022; 12:785375. [PMID: 34992536 PMCID: PMC8724575 DOI: 10.3389/fphar.2021.785375] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/30/2021] [Indexed: 12/29/2022] Open
Abstract
Chronic kidney disease (CKD) is one of the increasingly serious public health concerns worldwide; the global burden of CKD is increasingly due to high morbidity and mortality. At present, there are three key problems in the clinical treatment and management of CKD. First, the current diagnostic indicators, such as proteinuria and serum creatinine, are greatly interfered by the physiological conditions of patients, and the changes in the indicator level are not synchronized with renal damage. Second, the established diagnosis of suspected CKD still depends on biopsy, which is not suitable for contraindication patients, is also traumatic, and is not sensitive to early progression. Finally, the prognosis of CKD is affected by many factors; hence, it is ineviatble to develop effective biomarkers to predict CKD prognosis and improve the prognosis through early intervention. Accurate progression monitoring and prognosis improvement of CKD are extremely significant for improving the clinical treatment and management of CKD and reducing the social burden. Therefore, biomarkers reported in recent years, which could play important roles in accurate progression monitoring and prognosis improvement of CKD, were concluded and highlighted in this review article that aims to provide a reference for both the construction of CKD precision therapy system and the pharmaceutical research and development.
Collapse
Affiliation(s)
- Zhonghong Yan
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guanran Wang
- Heilongjiang University of Chinese Medicine, Harbin, China.,Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xingyang Shi
- Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
45
|
Du X, Wu J, Gao C, Tan Q, Xu Y. Effects of Resistant Starch on Patients with Chronic Kidney Disease: A Systematic Review and Meta-Analysis. J Diabetes Res 2022; 2022:1861009. [PMID: 35899018 PMCID: PMC9314005 DOI: 10.1155/2022/1861009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a main health problem associated with increased risk of cardiovascular disease, morbidity, and mortality. Recent studies shown that the progression of CKD may be related to the change of intestinal flora. Resistant starch (RS) is a type of dietary fiber that can act as a substrate for microbial fermentation. Some studies have found that the supplementation of RS can improve the intestinal flora disorder in CKD patients. However, the specific effect of RS on CKD patients remains controversial. OBJECTIVE We designed this meta-analysis to identify and assess the effects of RS on patients with CKD. METHODS A comprehensive search of MEDLINE, Embase, Web of Science, and Cochrane systematic review databases was conducted in January 2020, and all new trials were updated in August 2021. Randomized trials were collected to assess the effects of RS on patients with CKD. The weighted average effect size of the net change was calculated by using the random-effects model. RESULTS The meta-analysis included 8 studies involving 301 participants. RS intake significantly reduced serum indolephenol sulfate (IS), blood phosphorus, IL-6, and uric acid levels in dialysis patients. The mean difference (MD) of serum IS (P = 0.0002) in the dialysis subgroup was -12.57 μmol/L (95% CI: -19.28, -5.86 μmol/L). The MD of blood phosphorus (P = 0.03) was -0.39 mg/dl (95% CI: -0.78, -0.01 mg/dl). The MD of serum uric acid (P = 0.004) between the dialysis subgroup and the nondialysis subgroup was -31.58 mmol/L (95% CI: -52.99, -10.17 mmol/L). The mean difference (MD) of IL-6 (P = 0.02) in the dialysis subgroup was -1.16 μmol/L (95% CI: -2.16, -0.16 μmol/L). However, there was no significant change of RS on hs-CRP, serum creatinine, blood urea nitrogen (BUN), blood paracresol sulfate, and blood lipid. CONCLUSIONS The intake of RS reduced the serum IS, serum phosphorus, IL-6, and uric acid levels significantly in dialysis patients, while hs-CRP, serum creatinine, BUN, serum paracresol sulfate, and blood lipid showed no significant changes.
Collapse
Affiliation(s)
- Xinyi Du
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Wu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chenlin Gao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qinqin Tan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China 646000
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
46
|
Sobh E, Al-Adl A, Awadallah M, Abdelsalam K, Awad S, Surrati A, Alhadrami H. Coronavirus Disease-2019 and the kidneys: A tragedy of reciprocal damage and management challenges. JOURNAL OF MEDICAL SCIENCES 2022. [DOI: 10.4103/jmedsci.jmedsci_150_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
47
|
Scherberich JE, Fünfstück R, Naber KG. Urinary tract infections in patients with renal insufficiency and dialysis - epidemiology, pathogenesis, clinical symptoms, diagnosis and treatment. GMS INFECTIOUS DISEASES 2021; 9:Doc07. [PMID: 35106269 PMCID: PMC8777485 DOI: 10.3205/id000076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Epidemiological studies show an increasing number of patients worldwide suffering from chronic kidney diseases (CKD), which are associated with a risk for progression to end-stage kidney disease (ESKD). CKD patients stage 2-5, patients with regular chronic dialysis treatment (hemo- or peritoneal dialysis), and patients suffering from kidney allograft dysfunction are at high risk to develop infections, e.g. urinary tract infections (UTI) and/or sepsis (urosepsis). These groups show metabolic disturbance, chronic inflammation, and impaired immunocompetence. Escherichia coli is still the most common pathogen in UTI. A wide variety of other pathogens may be involved in UTI. Urological interventions, catheterization, as well as repeated courses of antibiotics contribute to an increased challenge of antimicrobial resistance. The diagnosis of UTI in CKD is based on standard clinical and laboratory criteria. Pyuria (≥10 leucocytes/µl) is more often observed in patients with oligoanuria and low bacterial colony counts. The treatment strategies for this population are based on the same principles as in patients with normal renal function. However, drugs cleared by the kidney or by dialysis membranes need dose adjustment. Antimicrobials with potential systemic toxicity and nephrotoxicity should be administered with caution.
Collapse
Affiliation(s)
| | | | - Kurt G. Naber
- Department of Urology, Technical University Munich, Germany
| |
Collapse
|
48
|
Merino-Ribas A, Araujo R, Bancu I, Graterol F, Vergara A, Noguera-Julian M, Paredes R, Bonal J, Sampaio-Maia B. Gut microbiome in hemodialysis patients treated with calcium acetate or treated with sucroferric oxyhydroxide: a pilot study. Int Urol Nephrol 2021; 54:2015-2023. [PMID: 34923600 PMCID: PMC9262763 DOI: 10.1007/s11255-021-03091-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/08/2021] [Indexed: 01/30/2023]
Abstract
PURPOSE It has been proved that the gut microbiome is altered in patients with chronic kidney disease. This contributes to chronic inflammation and increases cardiovascular risk and mortality, especially in those undergoing hemodialysis. Phosphate binders may potentially induce changes in their microbiome. This trial aimed to compare the changes in the gut microbiome of hemodialysis patients treated with calcium acetate to those treated with sucroferric oxyhydroxide. METHODS Twelve hemodialysis patients were distributed to receive calcium acetate or sucroferric oxyhydroxide for 5 months. Blood samples (for biochemical analysis) and stool samples (for microbiome analysis) were collected at baseline, 4, 12, and 20 weeks after treatment initiation. Fecal DNA was extracted and a 16S rRNA sequencing library was constructed targeting the V3 and V4 hypervariable regions. RESULTS Regarding clinical variables and laboratory parameters, no statistically significant differences were observed between calcium acetate or sucroferric oxyhydroxide groups. When analyzing stool samples, we found that all patients were different (p = 0.001) among themselves and these differences were kept along the 20 weeks of treatment. The clustering analysis in microbial profiles grouped the samples of the same patient independently of the treatment followed and the stage of the treatment. CONCLUSION These results suggest that a 5-month treatment with either calcium acetate or sucroferric oxyhydroxide did not modify baseline diversity or baseline bacterial composition in hemodialysis patients, also about the high-variability profiles of the gut microbiome found among these patients.
Collapse
Affiliation(s)
- Ana Merino-Ribas
- Universitat Autònoma de Barcelona, Barcelona, Spain. .,Nephrology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain. .,Nephrology Department, Hospital Universitari de Girona Doctor Josep Trueta, Avinguda de França S/N, 17007, Girona, Spain. .,i3S-Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Porto, Portugal.
| | - Ricardo Araujo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Porto, Portugal
| | - Ioana Bancu
- Universitat Autònoma de Barcelona, Barcelona, Spain.,Nephrology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Fredzzia Graterol
- Universitat Autònoma de Barcelona, Barcelona, Spain.,Nephrology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Andrea Vergara
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Jordi Bonal
- Nephrology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Benedita Sampaio-Maia
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Porto, Portugal.,Faculty of Dental Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
49
|
Matsui M, Tsuruya K, Yoshida H, Iseki K, Fujimoto S, Konta T, Moriyama T, Yamagata K, Narita I, Kasahara M, Shibagaki Y, Kondo M, Asahi K, Watanabe T. Trace proteinuria as a risk factor for cancer death in a general population. Sci Rep 2021; 11:16890. [PMID: 34413415 PMCID: PMC8376860 DOI: 10.1038/s41598-021-96388-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/06/2021] [Indexed: 11/29/2022] Open
Abstract
Growing evidence has demonstrated an association between nondialysis chronic kidney disease and cancer incidence, although the association between trace proteinuria and cancer death remains unclear. The aim of this study was to investigate the association between trace proteinuria and cancer death in a community-based population in Japan. This was a prospective cohort study of 377,202 adults who participated in the Japanese Specific Health Check and Guidance System from 2008 to 2011. Exposure was dipstick proteinuria categorized as − (negative), ± (trace), 1 + (mild), or ≥ 2 + (moderate to heavy). Outcome was cancer death based on information from the national database of death certificates. Adjusted Cox hazard regression model was used to evaluate the associations between trace proteinuria and cancer death. During median follow-up of 3.7 years, 3056 cancer deaths occurred, corresponding to overall cancer death rate of 21.7/10,000 person-years. In the fully adjusted model, risk of cancer death increased significantly in each successive category of proteinuria: hazard ratio (HR) (95% confidence interval [95% CI]) for risk of cancer death was 1.16 (1.03–1.31), 1.47 (1.27–1.70), and 1.61 (1.33–1.96) for trace, mild, and moderate to heavy proteinuria, respectively. Sensitivity analyses revealed a similar association between trace proteinuria and cancer death, and participants with trace proteinuria had greater risk of mortality from hematological cancers (HR: 1.59 [95% CI: 1.09–2.31]). Both mild to heavy and trace proteinuria were significantly associated with risk of mortality from cancer in a general population.
Collapse
Affiliation(s)
- Masaru Matsui
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan. .,Department of Nephrology, Nara Prefecture General Medical Center, 2-897-5 Shichijo-nishimachi, Nara, Japan.
| | - Kazuhiko Tsuruya
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan.,Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Hisako Yoshida
- Department of Medical Statistics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kunitoshi Iseki
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Shouichi Fujimoto
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Tsuneo Konta
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Toshiki Moriyama
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Kunihiro Yamagata
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Ichiei Narita
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Masato Kasahara
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Yugo Shibagaki
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Masahide Kondo
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Koichi Asahi
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| | - Tsuyoshi Watanabe
- Steering Committee of Research on Design of the Comprehensive Health Care System for Chronic Kidney Disease (CKD), Based on the Individual Risk Assessment by Specific Health Checkup, The Ministry of Health, Labour and Welfare of Japan, Fukushima, Japan
| |
Collapse
|
50
|
Karaduta O, Glazko G, Dvanajscak Z, Arthur J, Mackintosh S, Orr L, Rahmatallah Y, Yeruva L, Tackett A, Zybailov B. Resistant starch slows the progression of CKD in the 5/6 nephrectomy mouse model. Physiol Rep 2021; 8:e14610. [PMID: 33038060 PMCID: PMC7547583 DOI: 10.14814/phy2.14610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/31/2020] [Accepted: 09/21/2020] [Indexed: 01/02/2023] Open
Abstract
Background Resistant Starch (RS) improves CKD outcomes. In this report, we study how RS modulates host‐microbiome interactions in CKD by measuring changes in the abundance of proteins and bacteria in the gut. In addition, we demonstrate RS‐mediated reduction in CKD‐induced kidney damage. Methods Eight mice underwent 5/6 nephrectomy to induce CKD and eight served as healthy controls. CKD and Healthy (H) groups were further split into those receiving RS (CKDRS, n = 4; HRS, n = 4) and those on normal diet (CKD, n = 4, H, n = 4). Kidney injury was evaluated by measuring BUN/creatinine and by histopathological evaluation. Cecal contents were analyzed using mass spectrometry‐based metaproteomics and de novo sequencing using PEAKS. All the data were analyzed using R/Bioconductor packages. Results The 5/6 nephrectomy compromised kidney function as seen by an increase in BUN/creatinine compared to healthy groups. Histopathology of kidney sections showed reduced tubulointerstitial injury in the CKDRS versus CKD group; while no significant difference in BUN/creatinine was observed between the two CKD groups. Identified proteins point toward a higher population of butyrate‐producing bacteria, reduced abundance of mucin‐degrading bacteria in the RS fed groups, and to the downregulation of indole metabolism in CKD groups. Conclusion RS slows the progression of chronic kidney disease. Resistant starch supplementation leads to active bacterial proliferation and the reduction of harmful bacterial metabolites.
Collapse
Affiliation(s)
- Oleg Karaduta
- Department of Biochemistry and Molecular Biology, UAMS, Little Rock, AR, USA
| | - Galina Glazko
- Department of Biomedical Informatics, UAMS, Little Rock, AR, USA
| | | | - John Arthur
- Division of Nephrology, UAMS, Little Rock, AR, USA
| | - Samuel Mackintosh
- Department of Biochemistry and Molecular Biology, UAMS, Little Rock, AR, USA.,Proteomics Core Facility, UAMS, Little Rock, AR, USA
| | - Lisa Orr
- Department of Biochemistry and Molecular Biology, UAMS, Little Rock, AR, USA
| | | | - Laxmi Yeruva
- Department of Biochemistry and Molecular Biology, UAMS, Little Rock, AR, USA.,Arkansas Children's Nutrition Center, Little Rock, AR, USA.,Department of Pediatrics, UAMS, Little Rock, AR, USA
| | - Alan Tackett
- Department of Biochemistry and Molecular Biology, UAMS, Little Rock, AR, USA.,Proteomics Core Facility, UAMS, Little Rock, AR, USA.,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Boris Zybailov
- Department of Biochemistry and Molecular Biology, UAMS, Little Rock, AR, USA
| |
Collapse
|