1
|
Tong Y, Guo S, Li T, Yang K, Gao W, Peng F, Zou X. Gut microbiota and renal fibrosis. Life Sci 2024; 357:123072. [PMID: 39307181 DOI: 10.1016/j.lfs.2024.123072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Renal fibrosis represents a critical pathological condition in the progression of renal dysfunction, characterized by aberrant accumulation of extracellular matrix (ECM) and structural alterations in renal tissue. Recent research has highlighted the potential significance of gut microbiota and demonstrated their influence on host health and disease mechanisms through the production of bioactive metabolites. This review examines the role of alterations in gut microbial composition and their metabolites in the pathophysiological processes underlying renal fibrosis. It delineates current therapeutic interventions aimed at modulating gut microbiota composition, encompassing dietary modifications, pharmacological approaches, and probiotic supplementation, while evaluating their efficacy in mitigating renal fibrosis. Through a comprehensive analysis of current research findings, this review enhances our understanding of the bidirectional interaction between gut microbiota and renal fibrosis, establishing a theoretical foundation for future research directions and potential clinical applications in this domain.
Collapse
Affiliation(s)
- Yinghao Tong
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Shangze Guo
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Ting Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Kexin Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Fujun Peng
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
2
|
Si ZL, Wang HY, Wang T, Cao YZ, Li QZ, Liu K, Huang Z, Liu HL, Tan YJ, Wang YY, Huang FQ, Ma GX, Alolga RN, Yan M, Chen C, Li JH, Li J, Liu HW, Zhang ZH. Gut Bacteroides ovatus ameliorates renal fibrosis by promoting the production of HDCA through upregulation of Clostridium scindens. Cell Rep 2024; 43:114830. [PMID: 39392759 DOI: 10.1016/j.celrep.2024.114830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/14/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
Renal fibrosis, inflammation, and gut dysbiosis are all linked to chronic kidney disease (CKD). Here we show that Bacteroides ovatus protects against renal fibrosis. Mechanistically, B. ovatus enhances intestinal hyodeoxycholic acid (HDCA) levels by upregulating a strain of intestinal bacteria, Clostridium scindens, that has the capacity for direct HDCA production in mice. HDCA significantly promoted GLP-1 secretion by upregulating the expression of TGR5 and downregulating the expression of farnesoid X receptor (FXR) in the gut. Activation of renal GLP-1R attenuates renal fibrosis while delaying the subsequent development of CKD. In addition, HDCA can also protect against renal fibrosis by directly upregulating renal TGR5. The natural product neohesperidin (NHP) was found to exert its anti-renal fibrotic effects by promoting the growth of B. ovatus. Our findings provide mechanistic insights into the therapeutic potential of B. ovatus, C. scindens, and HDCA in treating CKD.
Collapse
Affiliation(s)
- Zi-Lin Si
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Han-Yu Wang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing 100101, P.R. China
| | - Yi-Zhi Cao
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qing-Zhen Li
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Kang Liu
- Department of Nephrology, Jiangsu Province Hospital (The First Affiliated Hospital of Nanjing Medical University), Nanjing 210029, China
| | - Zhou Huang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hui-Ling Liu
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Ya-Jie Tan
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yin-Yin Wang
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Feng-Qing Huang
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Gao-Xiang Ma
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Raphael N Alolga
- State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Miao Yan
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Cheng Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jun-Hui Li
- Putuo People's Hospital, Tongji University, Shanghai 200060, China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Hong-Wei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing 100101, P.R. China
| | - Zhi-Hao Zhang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCM Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
3
|
Rowe JC, Summers SC, Quimby JM, Winston JA. Fecal bile acid dysmetabolism and reduced ursodeoxycholic acid correlate with novel microbial signatures in feline chronic kidney disease. Front Microbiol 2024; 15:1458090. [PMID: 39498133 PMCID: PMC11532117 DOI: 10.3389/fmicb.2024.1458090] [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: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 11/07/2024] Open
Abstract
Background Microbial-derived secondary bile acids (SBAs) are reabsorbed and sensed via host receptors modulating cellular inflammation and fibrosis. Feline chronic kidney disease (CKD) occurs with progressive renal inflammation and fibrosis, mirroring the disease pathophysiology of human CKD patients. Methods Prospective cross-sectional study compared healthy cats (n = 6) with CKD (IRIS Stage 2 n = 17, Stage 3 or 4 n = 11). Single timepoint fecal samples from all cats underwent targeted bile acid metabolomics. 16S rRNA gene amplicon sequencing using DADA2 with SILVA taxonomy characterized the fecal microbiota. Results CKD cats had significantly reduced fecal concentrations (median 12.8 ng/mg, Mann-Whitney p = 0.0127) of the SBA ursodeoxycholic acid (UDCA) compared to healthy cats (median 39.4 ng/mg). Bile acid dysmetabolism characterized by <50% SBAs was present in 8/28 CKD and 0/6 healthy cats. Beta diversity significantly differed between cats with <50% SBAs and > 50% SBAs (PERMANOVA p < 0.0001). Twenty-six amplicon sequence variants (ASVs) with >97% nucleotide identity to Peptacetobacter hiranonis were identified. P. hiranonis combined relative abundance was significantly reduced (median 2.1%) in CKD cats with <50% SBAs compared to CKD cats with >50% SBAs (median 13.9%, adjusted p = 0.0002) and healthy cats with >50% SBAs (median 15.5%, adjusted p = 0.0112). P. hiranonis combined relative abundance was significantly positively correlated with the SBAs deoxycholic acid (Spearman r = 0.5218, adjusted p = 0.0407) and lithocholic acid (Spearman r = 0.5615, adjusted p = 0.0156). Three Oscillospirales ASVs and a Roseburia ASV were also identified as significantly correlated with fecal SBAs. Clinical and translational importance The gut-kidney axis mediated through microbial-derived SBAs appears relevant to the spontaneous animal CKD model of domestic cats. This includes reduced fecal concentrations of the microbial-derived SBA UDCA, known to regulate inflammation and fibrosis and be reno-protective. Microbes correlated with fecal SBAs include bai operon containing P. hiranonis, as well as members of Oscillospirales, which also harbor a functional bai operon. Ultimately, CKD cats represent a translational opportunity to study the role of SBAs in the gut-kidney axis, including the potential to identify novel microbial-directed therapeutics to mitigate CKD pathogenesis in veterinary patients and humans alike.
Collapse
Affiliation(s)
- John C. Rowe
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| | - Stacie C. Summers
- Department of Clinical Sciences, Oregon State University Carlson College of Veterinary Medicine, Corvallis, OR, United States
| | - Jessica M. Quimby
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| |
Collapse
|
4
|
Zhang W, Shen M, Chu P, Wang T, Ji J, Ning X, Yin S, Zhang K. Molecular characterization of CIRBP from Takifugu fasciatus and its potential roles in cold-induced liver damage. Int J Biol Macromol 2024; 281:136492. [PMID: 39393746 DOI: 10.1016/j.ijbiomac.2024.136492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
As a potent stressor, environmental cold stress induces severe mitochondrial dysfunction with the overproduction of reactive oxygen species (ROS) in fish, resulting in liver damage. However, the molecular mechanisms underlying the cold-induced liver damage remain unclear. In the present study, the cold-inducible RNA-binding protein (CIRBP) from Takifugu fasciatus was characterized, and its role in cold-induced oxidative stress damage was investigated. An acute liver injury model was constructed by exposing T. fasciatus individuals to temperatures of 25, 19, and 13 °C. Cold exposure markedly induced histomorphological liver injury and triggered endogenous apoptosis and NLRP3 inflammatory response. Cold treatment significantly increased CIRBP gene expression. A similar expression pattern was detected for thioredoxin (TRX), suggesting that these two proteins play a role in the establishment of cold adaptation. CIRBP binds directly to the 3'-UTR of TRX. Furthermore, in vivo experiment showed that, when CIRBP expression in T. fasciatus is knocked down, the time to loss equilibrium significantly shortened at 13 °C. Taken together, our study revealed that CIRBP is a critical protective factor against cold induced liver damage and that the CIRBP/TRX pathway could function as an underlying mechanism for cold adaptation in teleosts.
Collapse
Affiliation(s)
- Wenwen Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Minghao Shen
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Peng Chu
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China
| | - Tao Wang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China
| | - Jie Ji
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China
| | - Xianhui Ning
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China.
| | - Kai Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu 222005, China.
| |
Collapse
|
5
|
Zhang Q, Ye X, Zhu L, Xu Z, Hou Y, Ke Q, Feng J, Xie X, Chen D, Piao JG, Wei Y. Spatiotemporal delivery of multiple components of rhubarb-astragalus formula for the sysnergistic treatment of renal fibrosis. Front Pharmacol 2024; 15:1456721. [PMID: 39415839 PMCID: PMC11480027 DOI: 10.3389/fphar.2024.1456721] [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: 06/29/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose Rhubarb (Rheum palmatum L.) and astragalus (Radix astragali) find widespread used in clinical formulations for treating chronic kidney disease (CKD). Notably, the key active components, total rhubarb anthraquinone (TRA) and total astragalus saponin (TAS), exhibit superiority over rhubarb and astragalus in terms of their clear composition, stability, quality control, small dosage, and efficacy for disease treatment. Additionally, astragalus polysaccharides (APS) significantly contribute to the treatment of renal fibrosis by modulating the gut microbiota. However, due to differences in the biopharmaceutical properties of these components, achieving synergistic effects remains challenging. This study aims to develop combined pellets (CPs) and evaluate the potential effect on unilateral ureteral obstruction (UUO)-induced renal fibrosis. Methods The CPs pellets were obtained by combining TRA/TAS-loaded SNEDDS pellets and APS-loaded pellets, prepared using the fluidized bed coating process. The prepared pellets underwent evaluation for morphology, bulk density, hardness, and flowing property. Moreover, the in vitro release of the payloads was evaluated with the CHP Type I method. Furthermore, the unilateral ureteral obstruction (UUO) model was utilized to investigate the potential effects of CPs pellets on renal fibrosis and their contribution to gut microbiota modulation. Results The ex-vivo study demonstrated that the developed CPs pellets not only improved the dissolution of TRA and TAS but also delivered TRA/TAS and APS spatiotemporally to the appropriate site along the gastrointestinal tract. In an animal model of renal fibrosis (UUO rats), oral administration of the CPs ameliorated kidney histological pathology, reduced collagen deposition, and decreased the levels of inflammatory cytokines. The CPs also restored the disturbed gut microbiota induced by UUO surgery and protected the intestinal barrier. Conclusion The developed CPs pellets represent a promising strategy for efficiently delivering active components in traditional Chinese medicine formulas, offering an effective approach for treating CKD.
Collapse
Affiliation(s)
- Qibin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaofeng Ye
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhishi Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Hou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiaoying Ke
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiawei Feng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaowei Xie
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Danfei Chen
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Freitas ADS, Barroso FAL, Campos GM, Américo MF, Viegas RCDS, Gomes GC, Vital KD, Fernandes SOA, Carvalho RDDO, Jardin J, Miranda APGDS, Ferreira E, Martins FS, Laguna JG, Jan G, Azevedo V, de Jesus LCL. Exploring the anti-inflammatory effects of postbiotic proteins from Lactobacillus delbrueckii CIDCA 133 on inflammatory bowel disease model. Int J Biol Macromol 2024; 277:134216. [PMID: 39069058 DOI: 10.1016/j.ijbiomac.2024.134216] [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: 04/30/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Lactobacillus delbrueckii CIDCA 133 is a promising health-promoting bacterium shown to alleviate intestinal inflammation. However, the specific bacterial components responsible for these effects remain largely unknown. Here, we demonstrated that consuming extractable proteins from the CIDCA 133 strain effectively relieved acute ulcerative colitis in mice. This postbiotic protein fraction reduced the disease activity index and prevented colon shortening in mice. Furthermore, histological analysis revealed colitis prevention with reduced inflammatory cell infiltration into the colon mucosa. Postbiotic consumption also induced an immunomodulatory profile in colitic mice, as evidenced by both mRNA transcript levels (Tlr2, Nfkb1, Nlpr3, Tnf, and Il6) and cytokines concentration (IL1β, TGFβ, and IL10). Additionally, it enhanced the levels of secretory IgA, upregulated the transcript levels of tight junction proteins (Hp and F11r), and improved paracellular intestinal permeability. More interestingly, the consumption of postbiotic proteins modulated the gut microbiota (Bacteroides, Arkkemansia, Dorea, and Oscillospira). Pearson correlation analysis indicated that IL10 and IL1β levels were positively associated with Bacteroides and Arkkemansia_Lactobacillus abundance. Our study reveals that CIDCA 133-derived proteins possess anti-inflammatory properties in colonic inflammation.
Collapse
Affiliation(s)
- Andria Dos Santos Freitas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Gabriela Munis Campos
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Monique Ferrary Américo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Gabriel Camargos Gomes
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Kátia Duarte Vital
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | - Enio Ferreira
- Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Guimarães Laguna
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Vasco Azevedo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| | - Luís Cláudio Lima de Jesus
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
7
|
Xiao P, Hao J, Kuang Y, Dai C, Rong X, Jiang L, Xie Z, Zhang L, Chen Q, Liu E. Targeting Neuraminidase 4 Attenuates Kidney Fibrosis in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406936. [PMID: 39136142 PMCID: PMC11497051 DOI: 10.1002/advs.202406936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/01/2024] [Indexed: 10/25/2024]
Abstract
Despite significant progress in therapy, there remains a lack of substantial evidence regarding the molecular factors that lead to renal fibrosis. Neuraminidase 4 (NEU4), an enzyme that removes sialic acids from glycoconjugates, has an unclear role in chronic progressive fibrosis. Here, this study finds that NEU4 expression is markedly upregulated in mouse fibrotic kidneys induced by folic acid or unilateral ureter obstruction, and this elevation is observed in patients with renal fibrosis. NEU4 knockdown specifically in the kidney attenuates the epithelial-to-mesenchymal transition, reduces the production of pro-fibrotic cytokines, and decreases cellular senescence in male mice. Conversely, NEU4 overexpression exacerbates the progression of renal fibrosis. Mechanistically, NEU4254-388aa interacts with Yes-associated protein (YAP) at WW2 domain (231-263aa), promoting its nucleus translocation and activation of target genes, thereby contributing to renal fibrosis. 3,5,6,7,8,3',4'-Heptamethoxyflavone, a natural compound, is identified as a novel NEU4 inhibitor, effectively protecting mice from renal fibrosis in a NEU4-dependent manner. Collectively, the findings suggest that NEU4 may represent a promising therapeutic target for kidney fibrosis.
Collapse
Affiliation(s)
- Ping‐Ting Xiao
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing210009China
| | - Jin‐Hua Hao
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing210009China
| | - Yu‐Jia Kuang
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing210009China
| | - Cai‐Xia Dai
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
| | - Xiao‐Ling Rong
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
| | - Li‐Long Jiang
- PolyU Academy for Interdisciplinary ResearchThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Zhi‐Shen Xie
- Academy of Chinese Medical SciencesHenan University of Chinese MedicineZhengzhou450000China
| | - Lei Zhang
- Hunan Key Laboratory of Kidney Disease and Blood PurificationDepartment of NephrologyThe Second Xiangya Hospital Central South UniversityChangsha410000China
| | - Qian‐Qian Chen
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
| | - E‐Hu Liu
- School of PharmacyNanjing University of Chinese MedicineNanjing210023China
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing210009China
| |
Collapse
|
8
|
Zhang Z, Cao B, Wu Q. Causality of Genetically Determined Metabolites on Chronic Kidney Disease: A Two-Sample Mendelian Randomization Study In Silico. Metab Syndr Relat Disord 2024; 22:525-550. [PMID: 38742978 DOI: 10.1089/met.2024.0030] [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] [Indexed: 05/16/2024] Open
Abstract
Introduction: Chronic kidney disease (CKD) is associated with metabolic disorders. However, the evidence for the causality of circulating metabolites to promote or prevent CKD is still lacking. Methods: The two-sample Mendelian randomization (MR) analysis was conducted to evaluate the latent causal relationship between the genetically proxied 486 blood metabolites and CKD. Genome-wide association study (GWAS) data for exposures were derived from 7824 European GWAS on metabolite levels, which have been extensively utilized in the medical field to elucidate the mechanisms underlying disease onset and progression. The random inverse variance weighted (IVW) is the primary analysis for causality analysis while MR-Egger and weighted median as complementary analyses. For the further identification of metabolites, reverse MR and linkage disequilibrium score regression were performed for further evaluation. The drug target for N-acetylornithine was subsequently supplemented into the analysis, with MR and colocalization analysis being utilized. Key metabolic pathways were identified via MetaboAnalyst 4.0 (https://www.metaboanalyst.ca/) online website. Results: N-acetylornithine was identified as a reliable metabolite that increases the susceptibility to estimated glomerular filtration rate (eGFR) decrease (β = 0.047; 95% confidence interval: -0.068 to -0.026; PIVW = 1.5E-5). The "glyoxylate and dicarboxylate metabolism" pathway showed significant relevance to CKD development (P = 6E-4), whereas the "glycine, serine, and threonine metabolism" pathway was also recognized as associated with CKD by general practitioners (P = 7E-4). Colocalization analysis revealed a robust genetic link between N-acetylornithine and both CKD and eGFR, with 85.1% and 99.4% colocalization rates, respectively. IVW-MR analysis substantiated these findings with a significant positive association for CKD (odds ratio = 1.43, P = 4.7E-5) and a negative correlation with eGFR (b = -0.04, P = 1.13E-31). Conclusions: MR was utilized to explore the potential causal links between 61 genetic serum metabolites and CKD. N-acetylornithine and NAT8 were further explored as a potential therapeutic target for CKD treatment.
Collapse
Affiliation(s)
- Zekai Zhang
- Second College of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Beibei Cao
- Academy of Paediatrics, Nanjing Medical University, Nanjing, China
| | - Qiutong Wu
- Second College of Clinical Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Nan QY, Piao SG, Jin JZ, Chung BH, Yang CW, Li C. Pathogenesis and management of renal fibrosis induced by unilateral ureteral obstruction. Kidney Res Clin Pract 2024; 43:586-599. [PMID: 38325866 PMCID: PMC11467363 DOI: 10.23876/j.krcp.23.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 02/09/2024] Open
Abstract
Regardless of the underlying etiology, renal fibrosis is the final histological outcome of progressive kidney disease. Unilateral ureteral obstruction (UUO) is an ideal and reproducible experimental rodent model of renal fibrosis, which is characterized by tubulointerstitial inflammatory responses, accumulation of extracellular matrix, tubular dilatation and atrophy, and fibrosis. The magnitude of UUO-induced renal fibrosis is experimentally manipulated by the species chosen, animal age, and the severity and duration of the obstruction, while relief of the obstruction allows the animal to recover from fibrosis. The pathogenesis of renal fibrosis is complex and multifactorial and is orchestrated by activation of renin-angiotensin system (RAS), oxidative stress, inflammatory response, transforming growth factor beta 1-Smad pathway, activated myofibroblasts, cell death (apoptosis, autophagy, ferroptosis, and necroptosis), destruction of intracellular organelles, and signaling pathway. The current therapeutic approaches have limited efficacy. Inhibition of RAS and use of antioxidants and antidiabetic drugs, such as inhibitors of sodium-glucose cotransporter 2 and dipeptidyl peptidase-4, have recently gained attention as therapeutic strategies to prevent renal scarring. This literature review highlights the state of the art regarding the molecular mechanisms relevant to the management of renal fibrosis caused by UUO.
Collapse
Affiliation(s)
- Qi Yan Nan
- Department of Nephrology, Yanbian University Hospital, Yanji, China
- Department of Intensive Care Unit, Yanbian University Hospital, Yanji, China
| | - Shang Guo Piao
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Ji Zhe Jin
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Byung Ha Chung
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chul Woo Yang
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Can Li
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| |
Collapse
|
10
|
Huang Y, Cao J, Zhu M, Wang Z, Jin Z, Xiong Z. Nontoxigenic Bacteroides fragilis: A double-edged sword. Microbiol Res 2024; 286:127796. [PMID: 38870618 DOI: 10.1016/j.micres.2024.127796] [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: 09/23/2023] [Revised: 04/12/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
The contribution of commensal microbes to human health and disease is unknown. Bacteroides fragilis (B. fragilis) is an opportunistic pathogen and a common colonizer of the human gut. Nontoxigenic B. fragilis (NTBF) and enterotoxigenic B. fragilis (ETBF) are two kinds of B. fragilis. NTBF has been shown to affect the host immune system and interact with gut microbes and pathogenic microbes. Previous studies indicated that certain strains of B. fragilis have the potential to serve as probiotics, based on their observed relationship with the immune system. However, several recent studies have shown detrimental effects on the host when beneficial gut bacteria are found in the digestive system or elsewhere. In some pathological conditions, NTBF may have adverse reactions. This paper presents a comprehensive analysis of NTBF ecology from the host-microbe perspective, encompassing molecular disease mechanisms analysis, bacteria-bacteria interaction, bacteria-host interaction, and the intricate ecological context of the gut. Our review provides much-needed insights into the precise application of NTBF.
Collapse
Affiliation(s)
- Yumei Huang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiali Cao
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengpei Zhu
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ziwen Wang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ze Jin
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhifan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Li H, Du Y, Cheng K, Chen Y, Wei L, Pei Y, Wang X, Wang L, Zhang Y, Hu X, Lu Y, Zhu X. Gut microbiota-derived indole-3-acetic acid suppresses high myopia progression by promoting type I collagen synthesis. Cell Discov 2024; 10:89. [PMID: 39187483 PMCID: PMC11347609 DOI: 10.1038/s41421-024-00709-5] [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: 11/25/2023] [Accepted: 07/08/2024] [Indexed: 08/28/2024] Open
Abstract
High myopia (HM) is a leading cause of blindness worldwide with currently no effective interventions available. A major hurdle lies in its often isolated perception as a purely ocular morbidity, disregarding potential systemic implications. Recent evidence suggests the existence of a gut-eye axis; however, the role of gut microbiota in the pathogenesis of HM remains largely unexplored. Herein, we provide a potential crosstalk among HM's gut dysbiosis, microbial metabolites, and scleral remodeling. Utilizing 16S rRNA gene sequencing, we observed an altered gut microbiota profile in HM patients with a significant reduction in probiotic abundance compared with healthy controls. Subsequent targeted metabolic profiling revealed a notable decrease in plasma levels of the gut microbiota-derived metabolite indole-3-acetic acid (3-IAA) among HM patients, which is closely associated with the reduced probiotics, both negatively correlated with HM severity. Genetic analyses determined that gut microbiota are causally associated with myopia risk. Importantly, when mice subjected to HM modeling receive fecal microbiota transplantation from healthy donors, there is an increase in 3-IAA plasma levels and simultaneous retardation of HM progression along with better maintenance of collagen type I alpha 1 (COL1A1) expression in the sclera. Furthermore, 3-IAA gavage achieves similar effects. Mechanistic investigations confirm the transcriptional activation of COL1A1 by 3-IAA via promoting the enrichment of SP1 to its promoter. Together, our findings provide novel insights into the gut microbiota-eye axis in the pathogenesis of HM and propose new strategies for HM intervention by remodeling the gut microbiota and indole supplementation.
Collapse
Affiliation(s)
- Hao Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Du
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Kaiwen Cheng
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yuxi Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Ling Wei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yujun Pei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiaoyu Wang
- Mass Spectrometry Platform, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lan Wang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Ye Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiaoxin Hu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.
| | - Xiangjia Zhu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
- Key Laboratory of Myopia and Related Eye Diseases, NHC; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.
| |
Collapse
|
12
|
Xia J, Zhang Y, Zhang S, Lu C, Huan H, Guan X. Oat Dietary Fiber Delays the Progression of Chronic Kidney Disease in Mice by Modulating the Gut Microbiota and Reducing Uremic Toxin Levels. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38836841 DOI: 10.1021/acs.jafc.4c02591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Chronic kidney disease (CKD) has emerged as a significant public health concern. In this article, we investigated the mechanism of oat dietary fiber in regulating CKD. Our findings indicated that the gut microbiota of CKD patients promoted gut microbiota dysbiosis and kidney injury in CKD mice. Intervention with oat-resistant starch prepared by ultrasonic combined enzymatic hydrolysis (ORSU) and oat β-glucan with a molecular weight of 5 × 104 Da (OBGM) elevated the levels of short-chain fatty acids (SCFAs) and regulated gut dysbiosis in the gut-humanized CKD mice. ORSU and OBGM also reduced CKD-related uremic toxins such as creatinine, indoxyl sulfate (IS), and p-cresol sulfate (PCS) levels; reinforced the intestinal barrier function of the gut-humanized CKD mice; and mitigated renal inflammation and fibrosis via the NF-κB/TGF-β pathway. Therefore, ORSU and OBGM might delay the progression of CKD by modulating the gut microbiota to reduce uremic toxins levels. Our results explain the mechanism of oat dietary fiber aimed at mitigating CKD.
Collapse
Affiliation(s)
- Ji'an Xia
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Suhua Zhang
- Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu 215028, China
| | - Chunlai Lu
- The 905th Hospital of People's Liberation Army Navy, Shanghai 200050, China
| | - Hongdi Huan
- The 905th Hospital of People's Liberation Army Navy, Shanghai 200050, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| |
Collapse
|
13
|
Hou Y, Zhu L, Ye X, Ke Q, Zhang Q, Xie X, Piao JG, Wei Y. Integrated oral microgel system ameliorates renal fibrosis by hitchhiking co-delivery and targeted gut flora modulation. J Nanobiotechnology 2024; 22:305. [PMID: 38822364 PMCID: PMC11143587 DOI: 10.1186/s12951-024-02586-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Renal fibrosis is a progressive process associated with chronic kidney disease (CKD), contributing to impaired kidney function. Active constituents in traditional Chinese herbs, such as emodin (EMO) and asiatic acid (AA), exhibit potent anti-fibrotic properties. However, the oral administration of EMO and AA results in low bioavailability and limited kidney accumulation. Additionally, while oral probiotics have been accepted for CKD treatment through gut microbiota modulation, a significant challenge lies in ensuring their viability upon administration. Therefore, our study aims to address both renal fibrosis and gut microbiota imbalance through innovative co-delivery strategies. RESULTS In this study, we developed yeast cell wall particles (YCWPs) encapsulating EMO and AA self-assembled nanoparticles (NPYs) and embedded them, along with Lactobacillus casei Zhang, in chitosan/sodium alginate (CS/SA) microgels. The developed microgels showed significant controlled release properties for the loaded NPYs and prolonged the retention time of Lactobacillus casei Zhang (L. casei Zhang) in the intestine. Furthermore, in vivo biodistribution showed that the microgel-carried NPYs significantly accumulated in the obstructed kidneys of rats, thereby substantially increasing the accumulation of EMO and AA in the impaired kidneys. More importantly, through hitchhiking delivery based on yeast cell wall and positive modulation of gut microbiota, our microgels with this synergistic strategy of therapeutic and modulatory interactions could regulate the TGF-β/Smad signaling pathway and thus effectively ameliorate renal fibrosis in unilateral ureteral obstruction (UUO) rats. CONCLUSION In conclusion, our work provides a new strategy for the treatment of renal fibrosis based on hitchhiking co-delivery of nanodrugs and probiotics to achieve synergistic effects of disease treatment and targeted gut flora modulation.
Collapse
Affiliation(s)
- Yu Hou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaofeng Ye
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qiaoying Ke
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qibin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaowei Xie
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
14
|
de Jesus LCL, Freitas ADS, Dutra JDCF, Campos GM, Américo MF, Laguna JG, Dornelas EG, Carvalho RDDO, Vital KD, Fernandes SOA, Cardoso VN, de Oliveira JS, de Oliveira MFA, Faria AMC, Ferreira E, Souza RDO, Martins FS, Barroso FAL, Azevedo V. Lactobacillus delbrueckii CIDCA 133 fermented milk modulates inflammation and gut microbiota to alleviate acute colitis. Food Res Int 2024; 186:114322. [PMID: 38729712 DOI: 10.1016/j.foodres.2024.114322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024]
Abstract
Lactobacillus delbrueckii subsp. lactis CIDCA 133 is a health-promoting bacterium that can alleviate gut inflammation and improve the epithelial barrier in a mouse model of mucositis. Despite these beneficial effects, the protective potential of this strain in other inflammation models, such as inflammatory bowel disease, remains unexplored. Herein, we examined for the first time the efficacy of Lactobacillus delbrueckii CIDCA 133 incorporated into a fermented milk formulation in the recovery of inflammation, epithelial damage, and restoration of gut microbiota in mice with dextran sulfate sodium-induced colitis. Oral administration of Lactobacillus delbrueckii CIDCA 133 fermented milk relieved colitis by decreasing levels of inflammatory factors (myeloperoxidase, N-acetyl-β-D-glucosaminidase, toll-like receptor 2, nuclear factor-κB, interleukins 10 and 6, and tumor necrosis factor), secretory immunoglobulin A levels, and intestinal paracellular permeability. This immunobiotic also modulated the expression of tight junction proteins (zonulin and occludin) and the activation of short-chain fatty acids-related receptors (G-protein coupled receptors 43 and 109A). Colonic protection was effectively associated with acetate production and restoration of gut microbiota composition. Treatment with Lactobacillus delbrueckii CIDCA 133 fermented milk increased the abundance of Firmicutes members (Lactobacillus genus) while decreasing the abundance of Proteobacteria (Helicobacter genus) and Bacteroidetes members (Bacteroides genus). These promising outcomes influenced the mice's mucosal healing, colon length, body weight, and disease activity index, demonstrating that this immunobiotic could be explored as an alternative approach for managing inflammatory bowel disease.
Collapse
Affiliation(s)
- Luís Cláudio Lima de Jesus
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Andria Dos Santos Freitas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Joyce da Cruz Ferraz Dutra
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela Munis Campos
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Monique Ferrary Américo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Guimarães Laguna
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | - Evandro Gonçalves Dornelas
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil
| | | | - Kátia Duarte Vital
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | | | - Valbert Nascimento Cardoso
- Federal University of Minas Gerais, Department of Clinical and Toxicological Analysis, Belo Horizonte, Minas Gerais, Brazil
| | - Jamil Silvano de Oliveira
- Federal University of Minas Gerais, Department of Biochemistry and Immunology, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ana Maria Caetano Faria
- Federal University of Minas Gerais, Department of Biochemistry and Immunology, Belo Horizonte, Minas Gerais, Brazil
| | - Enio Ferreira
- Federal University of Minas Gerais, Department of General Pathology, Belo Horizonte, Minas Gerais, Brazil
| | - Ramon de Oliveira Souza
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil; Ezequiel Dias Foundation, Research and Development Board, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Federal University of Minas Gerais, Department of Microbiology, Belo Horizonte, Minas Gerais, Brazil
| | | | - Vasco Azevedo
- Federal University of Minas Gerais, Department of Genetics, Ecology, and Evolution, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
15
|
Guo B, Zhang W, Zhou Y, Zhang J, Zeng C, Sun P, Liu B. Fucoxanthin restructures the gut microbiota and metabolic functions of non-obese individuals in an in vitro fermentation model. Food Funct 2024; 15:4805-4817. [PMID: 38563411 DOI: 10.1039/d3fo05671f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Fucoxanthin, a carotenoid exclusively derived from algae, exerts its bioactivities with the modulation of the gut microbiota in mice. However, mechanisms through which fucoxanthin regulates the gut microbiota and its derived metabolites/metabolism in humans remain unclear. In this study, we investigated the effects of fucoxanthin on the gut microbiota and metabolism of non-obese individuals using an in vitro simulated digestion-fermentation cascade model. The results showed that about half of the fucoxanthin was not absorbed in the intestine, thus reaching the colon. The gut microbiota from fecal samples underwent significant changes after 48 or 72 hours in vitro fermentation. Specifically, fucoxanthin significantly enhanced the relative abundance of Bacteroidota and Parabacteroides, leading to improved functions of the gut microbiota in its development, glycan biosynthesis and metabolism as well as in improving the digestive system, endocrine system and immune system. The recovery of fucoxanthin during fermentation showed a decreasing trend with the slight bio-conversion of fucoxanthinol. Notably, fucoxanthin supplementation significantly altered metabolites, especially bile acids and indoles in the simulated human gut ecosystem. Correlation analysis indicated the involvement of the gut microbiota in the manipulation of these metabolites by fucoxanthin. Moreover, all these altered metabolites revealed the improvement in the capacity of fucoxanthin in manipulating gut metabolism, especially lipid metabolism. Overall, fucoxanthin determinedly reshaped the gut microbiota and metabolism, implying its potential health benefits in non-obese individuals.
Collapse
Affiliation(s)
- Bingbing Guo
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Weihao Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Yonghui Zhou
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Jingyi Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Chengchu Zeng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Peipei Sun
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bin Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China.
| |
Collapse
|
16
|
Chui ZSW, Chan LML, Zhang EWH, Liang S, Choi EPH, Lok KYW, Tun HM, Kwok JYY. Effects of microbiome-based interventions on neurodegenerative diseases: a systematic review and meta-analysis. Sci Rep 2024; 14:9558. [PMID: 38664425 PMCID: PMC11045862 DOI: 10.1038/s41598-024-59250-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are characterized by neuronal damage and progressive loss of neuron function. Microbiome-based interventions, such as dietary interventions, biotics, and fecal microbiome transplant, have been proposed as a novel approach to managing symptoms and modulating disease progression. Emerging clinical trials have investigated the efficacy of interventions modulating the GM in alleviating or reversing disease progression, yet no comprehensive synthesis have been done. A systematic review of the literature was therefore conducted to investigate the efficacy of microbiome-modulating methods. The search yielded 4051 articles, with 15 clinical trials included. The overall risk of bias was moderate in most studies. Most microbiome-modulating interventions changed the GM composition. Despite inconsistent changes in GM composition, the meta-analysis showed that microbiome-modulating interventions improved disease burden (SMD, - 0.57; 95% CI - 0.93 to - 0.21; I2 = 42%; P = 0.002) with a qualitative trend of improvement in constipation. However, current studies have high methodological heterogeneity and small sample sizes, requiring more well-designed and controlled studies to elucidate the complex linkage between microbiome, microbiome-modulating interventions, and NDDs.
Collapse
Affiliation(s)
- Zara Siu Wa Chui
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lily Man Lee Chan
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Esther Wan Hei Zhang
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Suisha Liang
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong SAR, China
| | - Edmond Pui Hang Choi
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kris Yuet Wan Lok
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hein Min Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jojo Yan Yan Kwok
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
- Centre on Behavioral Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
17
|
Zhang Y, Zhong W, Liu W, Wang X, Lin G, Lin J, Fang J, Mou X, Jiang S, Huang J, Zhao W, Zheng Z. Uncovering specific taxonomic and functional alteration of gut microbiota in chronic kidney disease through 16S rRNA data. Front Cell Infect Microbiol 2024; 14:1363276. [PMID: 38707511 PMCID: PMC11066246 DOI: 10.3389/fcimb.2024.1363276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Chronic kidney disease (CKD) is worldwide healthcare burden with growing incidence and death rate. Emerging evidence demonstrated the compositional and functional differences of gut microbiota in patients with CKD. As such, gut microbial features can be developed as diagnostic biomarkers and potential therapeutic target for CKD. Methods To eliminate the outcome bias arising from factors such as geographical distribution, sequencing platform, and data analysis techniques, we conducted a comprehensive analysis of the microbial differences between patients with CKD and healthy individuals based on multiple samples worldwide. A total of 980 samples from six references across three nations were incorporated from the PubMed, Web of Science, and GMrepo databases. The obtained 16S rRNA microbiome data were subjected to DADA2 processing, QIIME2 and PICRUSt2 analyses. Results The gut microbiota of patients with CKD differs significantly from that of healthy controls (HC), with a substantial decrease in the microbial diversity among the CKD group. Moreover, a significantly reduced abundance of bacteria Faecalibacterium prausnitzii (F. prausnitzii) was detected in the CKD group through linear discriminant analysis effect size (LEfSe) analysis, which may be associated with the alleviating effects against CKD. Notably, we identified CKD-depleted F. prausnitzii demonstrated a significant negative correlation with three pathways based on predictive functional analysis, suggesting its potential role in regulating systemic acidbase disturbance and pro-oxidant metabolism. Discussion Our findings demonstrated notable alterations of gut microbiota in CKD patients. Specific gut-beneficial microbiota, especially F. prausnitzii, may be developed as a preventive and therapeutic tool for CKD clinical management.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Weicong Zhong
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Wenting Liu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Xiaohua Wang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Gan Lin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Jiawen Lin
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Junxuan Fang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiangyu Mou
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Shan Jiang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jiayuan Huang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Wenjing Zhao
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
18
|
Huo D, Liang W, Wang D, Liu Q, Wang H, Wang Y, Zhang C, Cong C, Su X, Tan X, Zhang W, Han L, Zhang D, Wang M, Feng H. Roflupram alleviates autophagy defects and reduces mutant hSOD1-induced motor neuron damage in cell and mouse models of amyotrophic lateral sclerosis. Neuropharmacology 2024; 247:109812. [PMID: 38218579 DOI: 10.1016/j.neuropharm.2023.109812] [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: 06/27/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 01/15/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable disease involving motor neuron (MN) degeneration and is characterized by ongoing myasthenia and amyotrophia in adults. Most ALS patients die of respiratory muscle paralysis after an average of 3-5 years. Defective autophagy in MNs is considered an important trigger of ALS pathogenesis. Roflupram (ROF) was demonstrated to activate autophagy in microglial cells and exert protective effects against Parkinson's disease (PD) and Alzheimer's disease (AD). Therefore, our research aimed to investigate the efficacy and mechanism of ROF in treating ALS both in vivo and in vitro. We found that ROF could delay disease onset and prolong the survival of hSOD1-G93A transgenic mice. Moreover, ROF protected MNs in the anterior horn of the spinal cord, activated the AMPK/ULK1 signaling pathway, increased autophagic flow, and reduced SOD1 aggregation. In an NSC34 cell line stably transfected with hSOD1-G93A, ROF protected against cellular damage caused by hSOD1-G93A. Moreover, we have demonstrated that ROF inhibited gliosis in ALS model mice. Collectively, our study suggested that ROF is neuroprotective in ALS models and the AMPK/ULK1 signaling pathway is a potential therapeutic target in ALS, which increases autophagic flow and reduces SOD1 aggregation.
Collapse
Affiliation(s)
- Di Huo
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Weiwei Liang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Di Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Qiaochu Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Hongyong Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Ying Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Chunting Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei City, Anhui Province, PR China
| | - Chaohua Cong
- Department of Neurology, Shanghai JiaoTong University School of Medicine, Shanghai No. 9 People's Hospital, Shanghai, PR China
| | - Xiaoli Su
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Xingli Tan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Wenmo Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Ling Han
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Dongmei Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Ming Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Honglin Feng
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China.
| |
Collapse
|
19
|
Liu HL, Huang Z, Li QZ, Cao YZ, Wang HY, Alolgab RN, Deng XY, Zhang ZH. Schisandrin A alleviates renal fibrosis by inhibiting PKCβ and oxidative stress. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155372. [PMID: 38382281 DOI: 10.1016/j.phymed.2024.155372] [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: 09/17/2023] [Revised: 01/01/2024] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Renal fibrosis is a common pathway that drives the advancement of numerous kidney maladies towards end-stage kidney disease (ESKD). Suppressing renal fibrosis holds paramount clinical importance in forestalling or retarding the transition of chronic kidney diseases (CKD) to renal failure. Schisandrin A (Sch A) possesses renoprotective effect in acute kidney injury (AKI), but its effects on renal fibrosis and underlying mechanism(s) have not been studied. STUDY DESIGN Serum biochemical analysis, histological staining, and expression levels of related proteins were used to assess the effect of PKCβ knockdown on renal fibrosis progression. Untargeted metabolomics was used to assess the effect of PKCβ knockdown on serum metabolites. Unilateral Ureteral Obstruction (UUO) model and TGF-β induced HK-2 cells and NIH-3T3 cells were used to evaluate the effect of Schisandrin A (Sch A) on renal fibrosis. PKCβ overexpressed NIH-3T3 cells were used to verify the possible mechanism of Sch A. RESULTS PKCβ was upregulated in the UUO model. Knockdown of PKCβ mitigated the progression of renal fibrosis by ameliorating perturbations in serum metabolites and curbing oxidative stress. Sch A alleviated renal fibrosis by downregulating the expression of PKCβ in kidney. Treatment with Sch A significantly attenuated the upregulated proteins levels of FN, COL-I, PKCβ, Vimentin and α-SMA in UUO mice. Moreover, Sch A exhibited a beneficial impact on markers associated with oxidative stress, including MDA, SOD, and GSH-Px. Overexpression of PKCβ was found to counteract the renoprotective efficacy of Sch A in vitro. CONCLUSION Sch A alleviates renal fibrosis by inhibiting PKCβ and attenuating oxidative stress.
Collapse
Affiliation(s)
- Hui-Ling Liu
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhou Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qing-Zhen Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yi-Zhi Cao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Han-Yu Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Raphael N Alolgab
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xue-Yang Deng
- State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Zhi-Hao Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
20
|
Hu X, Gan L, Tang Z, Lin R, Liang Z, Li F, Zhu C, Han X, Zheng R, Shen J, Yu J, Luo N, Peng W, Tan J, Li X, Fan J, Wen Q, Wang X, Li J, Zheng X, Liu Q, Guo J, Shi G, Mao H, Chen W, Yin S, Zhou Y. A Natural Small Molecule Mitigates Kidney Fibrosis by Targeting Cdc42-mediated GSK-3β/β-catenin Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307850. [PMID: 38240457 PMCID: PMC10987128 DOI: 10.1002/advs.202307850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/08/2024] [Indexed: 04/04/2024]
Abstract
Kidney fibrosis is a common fate of chronic kidney diseases (CKDs), eventually leading to renal dysfunction. Yet, no effective treatment for this pathological process has been achieved. During the bioassay-guided chemical investigation of the medicinal plant Wikstroemia chamaedaphne, a daphne diterpenoid, daphnepedunin A (DA), is characterized as a promising anti-renal fibrotic lead. DA shows significant anti-kidney fibrosis effects in cultured renal fibroblasts and unilateral ureteral obstructed mice, being more potent than the clinical trial drug pirfenidone. Leveraging the thermal proteome profiling strategy, cell division cycle 42 (Cdc42) is identified as the direct target of DA. Mechanistically, DA targets to reduce Cdc42 activity and down-regulates its downstream phospho-protein kinase Cζ(p-PKCζ)/phospho-glycogen synthase kinase-3β (p-GSK-3β), thereby promoting β-catenin Ser33/37/Thr41 phosphorylation and ubiquitin-dependent proteolysis to block classical pro-fibrotic β-catenin signaling. These findings suggest that Cdc42 is a promising therapeutic target for kidney fibrosis, and highlight DA as a potent Cdc42 inhibitor for combating CKDs.
Collapse
Affiliation(s)
- Xinrong Hu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Lu Gan
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Ziwen Tang
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Ruoni Lin
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Zhou Liang
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Feng Li
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Changjian Zhu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Xu Han
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Ruilin Zheng
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Jiani Shen
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Jing Yu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Ning Luo
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Wenxing Peng
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Jiaqing Tan
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Xiaoyan Li
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Jinjin Fan
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Qiong Wen
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Xin Wang
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Jianbo Li
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Xunhua Zheng
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Qinghua Liu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Jianping Guo
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhou510080China
| | - Guo‐Ping Shi
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Haiping Mao
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Wei Chen
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| | - Sheng Yin
- School of Pharmaceutical SciencesSun Yat‐sen UniversityGuangzhou510006China
| | - Yi Zhou
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologySun Yat‐Sen UniversityGuangzhou510080China
| |
Collapse
|
21
|
Xing JH, Niu TM, Zou BS, Yang GL, Shi CW, Yan QS, Sun MJ, Yu T, Zhang SM, Feng XZ, Fan SH, Huang HB, Wang JH, Li MH, Jiang YL, Wang JZ, Cao X, Wang N, Zeng Y, Hu JT, Zhang D, Sun WS, Yang WT, Wang CF. Gut microbiota-derived LCA mediates the protective effect of PEDV infection in piglets. MICROBIOME 2024; 12:20. [PMID: 38317217 PMCID: PMC10840300 DOI: 10.1186/s40168-023-01734-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/30/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND The gut microbiota is a critical factor in the regulation of host health, but the relationship between the differential resistance of hosts to pathogens and the interaction of gut microbes is not yet clear. Herein, we investigated the potential correlation between the gut microbiota of piglets and their disease resistance using single-cell transcriptomics, 16S amplicon sequencing, metagenomics, and untargeted metabolomics. RESULTS Porcine epidemic diarrhea virus (PEDV) infection leads to significant changes in the gut microbiota of piglets. Notably, Landrace pigs lose their resistance quickly after being infected with PEDV, but transplanting the fecal microbiota of Min pigs to Landrace pigs alleviated the infection status. Macrogenomic and animal protection models identified Lactobacillus reuteri and Lactobacillus amylovorus in the gut microbiota as playing an anti-infective role. Moreover, metabolomic screening of the secondary bile acids' deoxycholic acid (DCA) and lithocholic acid (LCA) correlated significantly with Lactobacillus reuteri and Lactobacillus amylovorus, but only LCA exerted a protective function in the animal model. In addition, LCA supplementation altered the distribution of intestinal T-cell populations and resulted in significantly enriched CD8+ CTLs, and in vivo and in vitro experiments showed that LCA increased SLA-I expression in porcine intestinal epithelial cells via FXR receptors, thereby recruiting CD8+ CTLs to exert antiviral effects. CONCLUSIONS Overall, our findings indicate that the diversity of gut microbiota influences the development of the disease, and manipulating Lactobacillus reuteri and Lactobacillus amylovorus, as well as LCA, represents a promising strategy to improve PEDV infection in piglets. Video Abstract.
Collapse
Affiliation(s)
- Jun-Hong Xing
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Tian-Ming Niu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Bo-Shi Zou
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Qing-Song Yan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Ming-Jie Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Tong Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Shu-Min Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Xi-Ze Feng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Shu-Hui Fan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Jun-Hong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Ming-Han Li
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Jing-Tao Hu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Wu-Sheng Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| |
Collapse
|
22
|
Behrens F, Bartolomaeus H, Wilck N, Holle J. Gut-immune axis and cardiovascular risk in chronic kidney disease. Clin Kidney J 2024; 17:sfad303. [PMID: 38229879 PMCID: PMC10790347 DOI: 10.1093/ckj/sfad303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Indexed: 01/18/2024] Open
Abstract
Patients with chronic kidney disease (CKD) suffer from marked cardiovascular morbidity and mortality, so lowering the cardiovascular risk is paramount to improve quality of life and survival in CKD. Manifold mechanisms are hold accountable for the development of cardiovascular disease (CVD), and recently inflammation arose as novel risk factor significantly contributing to progression of CVD. While the gut microbiome was identified as key regulator of immunity and inflammation in several disease, CKD-related microbiome-immune interaction gains increasing importance. Here, we summarize the latest knowledge on microbiome dysbiosis in CKD, subsequent changes in bacterial and host metabolism and how this drives inflammation and CVD in CKD. Moreover, we outline potential therapeutic targets along the gut-immune-cardiovascular axis that could aid the combat of CVD development and high mortality in CKD.
Collapse
Affiliation(s)
- Felix Behrens
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology und Intensive Medical Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology und Intensive Medical Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| |
Collapse
|
23
|
Xie Z, Zhang M, Luo Y, Jin D, Guo X, Yang W, Zheng J, Zhang H, Zhang L, Deng C, Zheng W, Tan EK, Jin K, Zhu S, Wang Q. Healthy Human Fecal Microbiota Transplantation into Mice Attenuates MPTP-Induced Neurotoxicity via AMPK/SOD2 Pathway. Aging Dis 2023; 14:2193-2214. [PMID: 37199590 PMCID: PMC10676800 DOI: 10.14336/ad.2023.0309] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/09/2023] [Indexed: 05/19/2023] Open
Abstract
Increasing evidence has shown that gut dysbacteriosis may play a crucial role in neuroinflammation in Parkinson's disease (PD). However, the specific mechanisms that link gut microbiota to PD remain unexplored. Given the critical roles of blood-brain barrier (BBB) dysfunction and mitochondrial dysfunction in the development of PD, we aimed to evaluate the interactions among the gut microbiota, BBB, and mitochondrial resistance to oxidation and inflammation in PD. We investigated the effects of fecal microbiota transplantation (FMT) on the physiopathology of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. The aim was to explore the role of fecal microbiota from PD patients and healthy human controls in neuroinflammation, BBB components, and mitochondrial antioxidative capacity via the AMPK/SOD2 pathway. Compared to control mice, MPTP-treated mice exhibited elevated levels of Desulfovibrio, whereas mice given FMT from PD patients exhibited enriched levels of Akkermansia and mice given FMT from healthy humans showed no significant alterations in gut microbiota. Strikingly, FMT from PD patients to MPTP-treated mice significantly aggravated motor impairments, dopaminergic neurodegeneration, nigrostriatal glial activation and colonic inflammation, and inhibited the AMPK/SOD2 signaling pathway. However, FMT from healthy human controls greatly improved the aforementioned MPTP-caused effects. Surprisingly, the MPTP-treated mice displayed a significant loss in nigrostriatal pericytes, which was restored by FMT from healthy human controls. Our findings demonstrate that FMT from healthy human controls can correct gut dysbacteriosis and ameliorate neurodegeneration in the MPTP-induced PD mouse model by suppressing microgliosis and astrogliosis, ameliorating mitochondrial impairments via the AMPK/SOD2 pathway, and restoring the loss of nigrostriatal pericytes and BBB integrity. These findings raise the possibility that the alteration in the human gut microbiota may be a risk factor for PD and provide evidence for potential application of FMT in PD preclinical treatment.
Collapse
Affiliation(s)
- Zhenchao Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Mahui Zhang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Dana Jin
- College of Biological Science, University of California, Davis, CA 95616, USA.
| | - Xingfang Guo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Hongfei Zhang
- Department of Anaesthesiology, Zhujiang Hospital of Southern Medical University, Guangdong, China.
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Southern Medical University, Guangdong, China.
| | - Chao Deng
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, Australia.
| | - Wenhua Zheng
- Centre of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau, China.
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
Li Z, Zhang X, Wu H, Ma Z, Liu X, Ma J, Zhang D, Sheng L, Chen X, Zhang S. Hydrangea paniculata coumarins attenuate experimental membranous nephritis by bidirectional interactions with the gut microbiota. Commun Biol 2023; 6:1189. [PMID: 37993541 PMCID: PMC10665342 DOI: 10.1038/s42003-023-05581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023] Open
Abstract
Coumarins isolated from Hydrangea paniculata (HP) had a renal protective effect in experimental membranous nephritis (MN), but the mechanisms are not clear. Currently, we investigate whether the modulation of gut dysbiosis by HP contributes to its renal protection. Experimental MN rats were treated with HP for six weeks. Fecal 16S rDNA sequencing and metabolomics were performed. Fecal microbiota transplantation (FMT) was used for the evaluation study. The results demonstrate that deteriorated renal function and gut dysbiosis are found in MN rats, as manifested by a higher Firmicutes/Bacteroidetes ratio and reduced diversity and richness, but both changes were reversed by HP treatment. Reduced gut dysbiosis is correlated with improved colonic integrity and lower endotoxemia in HP-treated rats. HP normalized the abnormal level of fecal metabolites by increasing short-chain fatty acid production and hindering the production of uremic toxin precursors. FMT of HP-treated feces to MN animals moderately reduced endotoxemia and albuminuria. Moreover, major coumarins in HP were only biotransformed into more bioactive 7-hydroxycoumarin by gut microbiota, which strengthened the effect of HP in vivo. Depletion of the gut microbiota partially abolished its renal protective effect. In conclusion, the bidirectional interaction between HP and the gut microbiota contributes to its beneficial effect.
Collapse
Affiliation(s)
- Zhaojun Li
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Xingguang Zhang
- Department of Endocrinology, The seventh medical center of Chinese PLA General Hospital, Beijing, 100070, China
| | - Haijie Wu
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Zhiling Ma
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Xikun Liu
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Jie Ma
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Dongming Zhang
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China
| | - Li Sheng
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China.
| | - Xiaoguang Chen
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China.
| | - Sen Zhang
- State key laboratory of bioactive substances and functions of natural medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union medical college, Beijing, 100050, China.
| |
Collapse
|
25
|
Sun D, Herath J, Zhou S, Ellepola G, Meegaskumbura M. Associations of Batrachochytrium dendrobatidis with skin bacteria and fungi on Asian amphibian hosts. ISME COMMUNICATIONS 2023; 3:123. [PMID: 37993728 PMCID: PMC10665332 DOI: 10.1038/s43705-023-00332-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023]
Abstract
Amphibian skin harbors microorganisms that are associated with the fungal pathogen Batrachochytrium dendrobatidis (Bd), which causes chytridiomycosis, one of the most significant wildlife diseases known. This pathogen originated in Asia, where diverse Bd lineages exist; hence, native amphibian hosts have co-existed with Bd over long time periods. Determining the nuances of this co-existence is crucial for understanding the prevalence and spread of Bd from a microbial context. However, associations of Bd with the natural skin microbiome remain poorly understood for Asian hosts, especially in relation to skin-associated fungi. We used 16 S rRNA and fungal internal transcribed spacer (ITS) gene sequencing to characterize the skin microbiome of four native Asian amphibian species and examined the relationships between Bd infection and their skin bacterial and fungal communities; we also analyzed the correlates of the putative anti-Bd bacteria. We show that both skin bacterial and fungal community structure and composition had significant associations with infection status (Bd presence/absence) and infection intensity (frequency of Bd sequence reads). We also found that the putative anti-Bd bacterial richness was correlated with Bd infection status and infection intensity, and observed that the relative abundance of anti-Bd bacteria roughly correspond with changes in both Bd prevalence and mean infection intensity in populations. Additionally, the microbial co-occurrence network of infected frogs was significantly different from that of uninfected frogs that were characterized by more keystone nodes (connectors) and larger proportions in correlations between bacteria, suggesting stronger inter-module bacterial interactions. These results indicate that the mutual effects between Bd and skin-associated microbiome, including the interplay between bacteria and fungi, might vary with Bd infection in susceptible amphibian species. This knowledge will help in understanding the dynamics of Bd from a microbial perspective, potentially contributing to mitigate chytridiomycosis in other regions of the world.
Collapse
Affiliation(s)
- Dan Sun
- Guangxi Key Laboratory for Forest Ecology and Conservation, College of Forestry, Guangxi University, Nanning, Guangxi, 530000, People's Republic of China
| | - Jayampathi Herath
- Guangxi Key Laboratory for Forest Ecology and Conservation, College of Forestry, Guangxi University, Nanning, Guangxi, 530000, People's Republic of China
- School of Biomedical Sciences, International Institute of Health Sciences (IIHS), No 704 Negombo Rd, Welisara, 71722, Sri Lanka
| | - Shipeng Zhou
- Guangxi Key Laboratory for Forest Ecology and Conservation, College of Forestry, Guangxi University, Nanning, Guangxi, 530000, People's Republic of China
| | - Gajaba Ellepola
- Guangxi Key Laboratory for Forest Ecology and Conservation, College of Forestry, Guangxi University, Nanning, Guangxi, 530000, People's Republic of China
- Department of Zoology, Faculty of Science, University of Peradeniya, Peradeniya, KY20400, Sri Lanka
| | - Madhava Meegaskumbura
- Guangxi Key Laboratory for Forest Ecology and Conservation, College of Forestry, Guangxi University, Nanning, Guangxi, 530000, People's Republic of China.
| |
Collapse
|
26
|
陈 静, 黄 英, 赵 景. [Trimethylamine N-Oxide Induces Renal Fibrosis Through the PI3K/AKT/SREBP1 Pathway]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1105-1111. [PMID: 38162065 PMCID: PMC10752791 DOI: 10.12182/20231160106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Indexed: 01/03/2024]
Abstract
Objective To investigate the role and mechanism of trimethylamine N-oxide (TMAO), a uremic toxin, in renal fibrosis. Methods A total of 20 male BALB/c mice were randomly and evenly assigned to a Control group and a TMAO group. Mice in the Control group received intraperitoneal injection of normal saline, while mice in the TMAO group received intraperitoneal injection of TMAO (20 mg/[kg·d]). The injection was given once a day for 8 weeks. Histopathology and fibrosis of kidney were observed by H&E staining and Masson staining. Immunohistochemistry was performed to determine the levels of alpha smooth muscle actin (α-SMA), recombinant human fibronectin fragment (Fibronectin), and sterol-regulatory element binding protein 1 (SREBP1). Western blot was performed to determine α-SMA, SREBP1, phosphatidylinositol 3 kinase (PI3K), phospho-phosphatidylinositol 3 kinase (p-PI3K), protein kinase B (PKB, also known as AKT), and phospho-AKT (p-AKT) protein levels. HK2 cells were treated with SREBP1 small interfering RNA (siRNA) and PI3K/AKT inhibitor, respectively, and the reversal of the effects of TMAO was examined. Results Animal experiments showed that, compared with the Control group, the mice treated with TMAO experienced pathological damage and fibrosis of the kidney tissue and the expression levels of fibrosis markers, α-SMA and Fibronectin, in the kidney were increased (all P<0.05). According to the findings from further investigation, the TMAO-treatment group showed increased expression of SREBP1 and an up-regulation of PI3K phosphorylation ratio and AKT phosphorylation ratio compared with those of the Control group (all P<0.05). Cell experiments produced results similar to those of the animal experiment. After siRNA interference with SREBP1 expression, the expression levels of fibrosis marker proteins decreased (P<0.05). Besides, the high expression of SREBP1 caused by TMAO was inhibited after HK2 cells were incubated with LY294002, a PI3K-AKT pathway inhibitor (P<0.05). Conclusion TMAO may induce renal fibrosis by promoting the PI3K/AKT/SREBP1 pathway.
Collapse
Affiliation(s)
- 静 陈
- 陆军军医大学第二附属医院 肾脏内科 (重庆 400037)Department of Nephrology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - 英辉 黄
- 陆军军医大学第二附属医院 肾脏内科 (重庆 400037)Department of Nephrology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - 景宏 赵
- 陆军军医大学第二附属医院 肾脏内科 (重庆 400037)Department of Nephrology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
27
|
Zhang C, Sui Y, Liu S, Yang M. Molecular mechanisms of metabolic disease-associated hepatic inflammation in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. EXPLORATION OF DIGESTIVE DISEASES 2023:246-275. [DOI: https:/doi.org/10.37349/edd.2023.00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/05/2023] [Indexed: 11/27/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the leading chronic liver disease worldwide, with a progressive form of non-alcoholic steatohepatitis (NASH). It may progress to advanced liver diseases, including liver fibrosis, cirrhosis, and hepatocellular carcinoma. NAFLD/NASH is a comorbidity of many metabolic disorders such as obesity, insulin resistance, type 2 diabetes, cardiovascular disease, and chronic kidney disease. These metabolic diseases are often accompanied by systemic or extrahepatic inflammation, which plays an important role in the pathogenesis and treatment of NAFLD or NASH. Metabolites, such as short-chain fatty acids, impact the function, inflammation, and death of hepatocytes, the primary parenchymal cells in the liver tissue. Cholangiocytes, the epithelial cells that line the bile ducts, can differentiate into proliferative hepatocytes in chronic liver injury. In addition, hepatic non-parenchymal cells, including liver sinusoidal endothelial cells, hepatic stellate cells, and innate and adaptive immune cells, are involved in liver inflammation. Proteins such as fibroblast growth factors, acetyl-coenzyme A carboxylases, and nuclear factor erythroid 2-related factor 2 are involved in liver metabolism and inflammation, which are potential targets for NASH treatment. This review focuses on the effects of metabolic disease-induced extrahepatic inflammation, liver inflammation, and the cellular and molecular mechanisms of liver metabolism on the development and progression of NAFLD and NASH, as well as the associated treatments.
Collapse
Affiliation(s)
- Chunye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Yuxiang Sui
- School of Life Science, Shanxi Normal University, Linfen 041004, Shanxi Province, China
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
28
|
Yang J, Meng L, Li Y, Huang H. Strategies for applying probiotics in the antibiotic management of Clostridioides difficile infection. Food Funct 2023; 14:8711-8733. [PMID: 37725066 DOI: 10.1039/d3fo02110f] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
The vital role of probiotics in the food field has been widely recognized, and at the same time, probiotics are gradually exhibiting surprising effects in the field of nutraceuticals, especially in regulating gut inflammation and the nutritional environment. As a dietary supplement in clinical nutrition, the coadministration of probiotics with antibiotics model has been applied to prevent intestinal infections caused by Clostridioides difficile. However, the mechanism behind this "bacteria-drug combination" model remains unclear. In particular, the selection of specific probiotic strains, the order of probiotics or antibiotics, and the time interval of coadministration are key issues that need to be further explored and clarified. Here, we focus on the issues mentioned above and give reasonable opinions, mainly including: (1) probiotics are safer and more effective when they intervene after antibiotics have been used; (2) the choice of the time interval between coadministration should be based on the metabolism of antibiotics in the host, differences in probiotic strains, the baseline ecological environment of the host's intestine, and the host immune level; in addition, the selection of the coadministration regime should also take into account factors such as the antibiotic sensitivity of probiotics and dosage of probiotics; and (3) by encapsulating probiotics, combining probiotics with prebiotics, and developing next-generation probiotics (NGPs) and postbiotic formulations, we can provide a more reasonable reference for this type of "bacteria-drug combination" model, and also provide targeted guidance for the application of probiotic dietary supplements in the antibiotic management of C. difficile infection.
Collapse
Affiliation(s)
- Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, China.
| | - Lingtong Meng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, China.
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, 2 Xuelin Road, Qixia District, Nanjing, China.
| |
Collapse
|
29
|
Wang J, Guo X, Zou Z, Yu M, Li X, Xu H, Chen Y, Jiao T, Wang K, Ma Y, Jiang J, Liang X, Wang J, Xie C, Zhong Y. Ootheca mantidis mitigates renal fibrosis in mice by the suppression of apoptosis via increasing the gut microbe Akkermansia muciniphila and modulating glutamine metabolism. Biomed Pharmacother 2023; 166:115434. [PMID: 37677965 DOI: 10.1016/j.biopha.2023.115434] [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: 07/01/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
Renal interstitial fibrosis (RIF), a progressive process affecting the kidneys in chronic kidney disease (CKD), currently lacks an effective therapeutic intervention. Traditional Chinese medicine (TCM) has shown promise in reducing RIF and slowing CKD progression. In this study, we demonstrated the dose-dependent attenuation of RIF by Ootheca mantidis (SPX), a commonly prescribed TCM for CKD, in a mouse model of unilateral ureteral obstruction (UUO). RNA-sequencing analysis suggested that SPX treatment prominently downregulated apoptosis and inflammation-associated pathways, thereby inhibiting the fibrogenic signaling in the kidney. We further found that transplantation of fecal microbiota from SPX-treated mice conferred protection against renal injury and fibrosis through suppressing apoptosis in UUO mice, indicating that SPX ameliorated RIF via remodeling the gut microbiota and reducing apoptosis in the kidneys. Further functional exploration of the gut microbiota combined with fecal metabolomics revealed increased levels of some probiotics, including Akkermansia muciniphila (A. muciniphila), and modulations in glutamine-related amino acid metabolism in UUO mice treated with SPX. Subsequent colonization of A. muciniphila and supplementation with glutamine effectively mitigated cell apoptosis and RIF in UUO mice. Collectively, these findings unveil a functionally A. muciniphila- and glutamine-involved gut-renal axis that contributes to the action of SPX, and provide important clue for the therapeutic potential of SPX, A. muciniphila, and glutamine in combatting RIF.
Collapse
Affiliation(s)
- Jue Wang
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Ziyuan Zou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Minjun Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Xueling Li
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Hualing Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yiping Chen
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Tingying Jiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China.
| | - Kanglong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Yuandi Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Jie Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xinyu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jiawen Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Yifei Zhong
- Division of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
30
|
Feng S, Liu Y, Xu J, Fan J, Li J, Wu Z, Sun Y, Xiong W. Three Strains of Lactobacillus Derived from Piglets Alleviated Intestinal Oxidative Stress Induced by Diquat through Extracellular Vesicles. Nutrients 2023; 15:4198. [PMID: 37836484 PMCID: PMC10574712 DOI: 10.3390/nu15194198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/04/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Previous studies found that Poria cocos polysaccharides (PCPs) significantly enhanced the antioxidant activity in piglet intestines while increasing the abundance of Lactobacillus. However, the relationship between Lactobacillus and antioxidant activity has yet to be verified, and the mode of action needs further investigation. Six Lactobacillus strains isolated from the intestines of neonatal piglets fed with PCPs were studied to investigate the relationship between Lactobacillus and intestinal oxidative stress. The results showed that three of them alleviated intestinal oxidative stress and protected the intestinal barrier. Subsequently, we extracted the extracellular vesicles (EVs) of these three Lactobacillus strains to verify their intestinal protection mode of action. We found that these EVs exerted an excellent antioxidant effect and intestinal barrier protection and could directly improve intestinal microbial composition. Our findings suggested that the EVs of the three Lactobacillus strains could enhance antioxidant activity by improving the physical intestinal barrier and remodeling gut microbiota. Unlike probiotics, which should be pre-colonized, EVs can act directly on the intestines. This study provides new ideas for the subsequent development of products to protect intestinal health.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wen Xiong
- College of Animal Sciences and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (S.F.); (Y.L.); (J.X.); (J.F.); (J.L.); (Z.W.); (Y.S.)
| |
Collapse
|
31
|
Lin Z, Li S, Xiao H, Xu Z, Li C, Zeng J, Wang S, Liu Z, Huang H. The degradation of TGR5 mediated by Smurf1 contributes to diabetic nephropathy. Cell Rep 2023; 42:112851. [PMID: 37481723 DOI: 10.1016/j.celrep.2023.112851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/07/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023] Open
Abstract
The multiple roles of TGR5 in the regulation of glucose metabolism, inflammation, and oxidative stress have drawn attention as therapeutic candidates for diabetes-related kidney disease. However, diabetes induces downregulation of renal TGR5 protein expression, and the regulatory mechanisms have not been clarified. Here, we identify that Smurf1, an E3 ubiquitin ligase, is a critical interactor of TGR5 and mediates the ubiquitination and proteasomal degradation of TGR5 under high glucose stimulation in glomerular mesangial cells. Genetic deficiency of Smurf1 restores TGR5 protein expression and attenuates renal injuries in diabetic mice. Mechanistically, Smurf1 interacts with the TGR5 ICL2 region by its HECT domain and induces K11/K48-linked polyubiquitination of TGR5 at K306 residue. Moreover, restoration of TGR5 protects db/db mice from diabetic nephropathy. These observations elucidate the critical role of Smurf1 in regulating TGR5 stability, suggesting that pharmacological targeting of the interaction between Smurf1 and TGR5 could serve as a promising therapeutic strategy against diabetic nephropathy.
Collapse
Affiliation(s)
- Zeyuan Lin
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhanchi Xu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chuting Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jingran Zeng
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shaogui Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Zhongqiu Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
32
|
Orozco-Aguilar J, Tacchi F, Aguirre F, Valero-Breton M, Castro-Sepulveda M, Simon F, Cabello-Verrugio C. Ursodeoxycholic acid induces sarcopenia associated with decreased protein synthesis and autophagic flux. Biol Res 2023; 56:28. [PMID: 37237400 DOI: 10.1186/s40659-023-00431-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/13/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Skeletal muscle generates force and movements and maintains posture. Under pathological conditions, muscle fibers suffer an imbalance in protein synthesis/degradation. This event causes muscle mass loss and decreased strength and muscle function, a syndrome known as sarcopenia. Recently, our laboratory described secondary sarcopenia in a chronic cholestatic liver disease (CCLD) mouse model. Interestingly, the administration of ursodeoxycholic acid (UDCA), a hydrophilic bile acid, is an effective therapy for cholestatic hepatic alterations. However, the effect of UDCA on skeletal muscle mass and functionality has never been evaluated, nor the possible involved mechanisms. METHODS We assessed the ability of UDCA to generate sarcopenia in C57BL6 mice and develop a sarcopenic-like phenotype in C2C12 myotubes and isolated muscle fibers. In mice, we measured muscle strength by a grip strength test, muscle mass by bioimpedance and mass for specific muscles, and physical function by a treadmill test. We also detected the fiber's diameter and content of sarcomeric proteins. In C2C12 myotubes and/or isolated muscle fibers, we determined the diameter and troponin I level to validate the cellular effect. Moreover, to evaluate possible mechanisms, we detected puromycin incorporation, p70S6K, and 4EBP1 to evaluate protein synthesis and ULK1, LC3 I, and II protein levels to determine autophagic flux. The mitophagosome-like structures were detected by transmission electron microscopy. RESULTS UDCA induced sarcopenia in healthy mice, evidenced by decreased strength, muscle mass, and physical function, with a decline in the fiber's diameter and the troponin I protein levels. In the C2C12 myotubes, we observed that UDCA caused a reduction in the diameter and content of MHC, troponin I, puromycin incorporation, and phosphorylated forms of p70S6K and 4EBP1. Further, we detected increased levels of phosphorylated ULK1, the LC3II/LC3I ratio, and the number of mitophagosome-like structures. These data suggest that UDCA induces a sarcopenic-like phenotype with decreased protein synthesis and autophagic flux. CONCLUSIONS Our results indicate that UDCA induces sarcopenia in mice and sarcopenic-like features in C2C12 myotubes and/or isolated muscle fibers concomitantly with decreased protein synthesis and alterations in autophagic flux.
Collapse
Affiliation(s)
- Josué Orozco-Aguilar
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute On Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Franco Tacchi
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute On Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Francisco Aguirre
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute On Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Mayalen Valero-Breton
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute On Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Mauricio Castro-Sepulveda
- Exercise Physiology and Metabolism Laboratory, School of Kinesiology, Faculty of Medicine, Finis Terrae University, Santiago, Chile
| | - Felipe Simon
- Millennium Institute On Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile.
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute On Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
33
|
Liu Y, Lei S, Hou R, Li D, Wan X, Cai H, Chen G. Tea polysaccharides from Taiping Houkui may serve as a potential candidate for regulation of lipid metabolism: Roles of gut microbiota and metabolite in vitro. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
|
34
|
Wilson NG, Hernandez-Leyva A, Rosen AL, Jaeger N, McDonough RT, Santiago-Borges J, Lint MA, Rosen TR, Tomera CP, Bacharier LB, Swamidass SJ, Kau AL. The gut microbiota of people with asthma influences lung inflammation in gnotobiotic mice. iScience 2023; 26:105991. [PMID: 36824270 PMCID: PMC9941210 DOI: 10.1016/j.isci.2023.105991] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The gut microbiota in early childhood is linked to asthma risk, but may continue to affect older patients with asthma. Here, we profile the gut microbiota of 38 children (19 asthma, median age 8) and 57 adults (17 asthma, median age 28) by 16S rRNA sequencing and find individuals with asthma harbored compositional differences from healthy controls in both adults and children. We develop a model to aid the design of mechanistic experiments in gnotobiotic mice and show enterotoxigenic Bacteroides fragilis (ETBF) is more prevalent in the gut microbiota of patients with asthma compared to healthy controls. In mice, ETBF, modulated by community context, can increase oxidative stress in the lungs during allergic airway inflammation (AAI). Our results provide evidence that ETBF affects the phenotype of airway inflammation in a subset of patients with asthma which suggests that therapies targeting the gut microbiota may be helpful tools for asthma control.
Collapse
Affiliation(s)
- Naomi G. Wilson
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ariel Hernandez-Leyva
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne L. Rosen
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natalia Jaeger
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan T. McDonough
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jesus Santiago-Borges
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael A. Lint
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas R. Rosen
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher P. Tomera
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leonard B. Bacharier
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - S. Joshua Swamidass
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew L. Kau
- Division of Allergy and Immunology, Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
35
|
Gao Q, Luo X. Research Highlights. Transplantation 2023; 107:6-7. [PMID: 37779320 DOI: 10.1097/tp.0000000000004485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Qimeng Gao
- Department of Surgery, Duke University Medical Center, Durham, NC
| | - Xunrong Luo
- Department of Medicine, Division of Nephrology, Duke University Medical Center, Durham, NC
| |
Collapse
|