101
|
Lamacchia O, Menzaghi C, Copetti M, Mastroianno M, Corsano C, Prehn C, Adamski J, Fontana A, Trischitta V, De Cosmo S. GFR Decline Predicts Total Mortality and Mediates the Effect of Tryptophan Metabolism on Death Risk in Type 2 Diabetes. J Clin Endocrinol Metab 2025; 110:e1451-e1457. [PMID: 39136240 DOI: 10.1210/clinem/dgae551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Indexed: 04/24/2025]
Abstract
CONTEXT The independent role of glomerular filtration rate (GFR) decline in shaping the risk of mortality in people with type 2 diabetes has only been partially addressed. OBJECTIVE The objective of the study was 2-fold: (1) to investigate the association between all-cause mortality and eGFR changes over time; (2) to understand whether renal dysfunction mediates the effect of tryptophan metabolism on death risk. METHODS Prospective study with an average follow-up of 14.8 years at a research hospital. The aggregate Gargano Mortality Study included 962 patients with type 2 diabetes who had at least 3 eGFR recordings and at least 1.5 years of follow-up. This was an observational study, with no interventions. Rate of all-cause mortality was measured. RESULTS Age- and sex-adjusted annual incident rate of mortality was 2.75 events per 100 person-years. The median annual rate of decline of eGFR was 1.3 mL/min per 1.73 m2 per year (range -3.7; 7.8). The decline of kidney function was strongly and independently associated with the risk of death. Serum kynurenine to tryptophan ratio (KTR) was associated with both eGFR decline and all-cause mortality. Causal mediation analysis showed that 24.3% of the association between KTR and mortality was mediated by eGFR decline. CONCLUSION In patients with type 2 diabetes, eGFR decline is independently associated with the risk of all-cause mortality and mediates a significant proportion of the association between tryptophan metabolism and death.
Collapse
Affiliation(s)
- Olga Lamacchia
- Department of Medical and Surgical Sciences, Endocrinology Unit, University of Foggia, 71122 Foggia, Italy
| | - Claudia Menzaghi
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Mario Mastroianno
- Scientific Direction, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Chiara Corsano
- Department of Medical and Surgical Sciences, Endocrinology Unit, University of Foggia, 71122 Foggia, Italy
- Department of Medical and Surgical Sciences, Hygiene Unit, University of Foggia, 71122 Foggia, Italy
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Andrea Fontana
- Unit of Biostatistics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Salvatore De Cosmo
- Unit of Internal Medicine, IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
102
|
Lyu Y, Zhang T, Zhong W, Yi S, Zhu L. Exposure to Sodium p-Perfluorous Nonenoxybenzenesulfonate Induces Renal Fibrosis in Mice by Disrupting Lysine Metabolism. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:7461-7473. [PMID: 40116701 DOI: 10.1021/acs.est.4c10724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Environmental exposure is one driving factor of chronic kidney disease (CKD), yet the intrinsic molecular mechanisms are largely unexplored. As a persistent chemical, perfluorooctanesulfonate (PFOS) is regulated due to a great potential to induce multiple diseases, including renal fibrosis, a major pathological characteristic of CKD. It is hypothesized that sodium p-perfluorous nonenoxybenzenesulfonate (OBS), a typical alternative to PFOS, may also induce renal fibrosis. We observed distinct renal fibrosis in mice exposed to OBS. Metabolomics analysis showed that Nα-acetyllysine was the primary metabolite biomarker, whose level decreased greatly due to its excessive consumption by lysyloxidase (LOX). This suppressed the miR-140-5p expression, promoting upregulation of fibroblast growth factor 9 (FGF9), which activated the PI3K/Akt signaling pathway through fibroblast growth factor receptor 3 (FGFR3), thereby enhancing proliferation and activation of fibroblasts. Supplement of Nα-acetyllysine upregulated miR-140-5p expression, reduced expressions of FGF9 and FGFR3, and eventually ameliorated OBS-induced renal fibrosis. Similarly, treatment with miR-140-5p agomir and PI3K/Akt signaling pathway inhibitor LY294002 attenuated OBS-induced renal fibrosis. Taken together, OBS caused renal fibrosis through the LOX-Nα-acetyllysine-miR-140-5p-FGF9-FGFR3-PI3K/Akt-Bad-Bcl-2-fibroblast axis. The results of this study reveal a specific molecular axis for OBS to induce renal fibrosis and call for concerns in supervising the application of OBS.
Collapse
Affiliation(s)
- Yang Lyu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Tianxu Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Wenjue Zhong
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Shujun Yi
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| |
Collapse
|
103
|
Tian J, Yang X, Fan S, Peng X, Su H, Bi H, Qiu M. Highly oxygenated lanostane triterpenoids from Ganoderma applanatum and their anti-liver fibrosis effects. Bioorg Chem 2025; 161:108497. [PMID: 40288012 DOI: 10.1016/j.bioorg.2025.108497] [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: 12/15/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Inspired by the intriguing structures and significant activities of Ganoderma triterpenoids (GTs), the EtOAc extract of Ganoderma applanatum was phytochemically investigated, leading to the isolation of 11 GTs, including 10 new one (1-10). Their structures including absolute configurations, were elucidated through IR, UV, HRESIMS, 1D NMR and 2D NMR data analyses. Notably, applanoids J (1) and K (2) represent the first example of GTs with an unprecedented B-seco-lanostane architecture featuring 1,4-cyclohexanedione motif. Meanwhile, compounds 1-7 and 9-11 were evaluated for their hepatoprotective activity using TGF-β1-induced liver fibrosis model, and some of them such as compounds 2, 4, 6, 7 and 10 significantly suppressed the abnormal upregulation of fibrosis-related genes FN, ACTA2 (encode α-SMA) and COL1A1. Mechanistic studies suggested that the anti-liver fibrosis effect of compound 7 may be mediated through inhibition of the TGF-β/Smad signaling pathway. This study not only illustrates the structural diversity of GTs but also highlights their potential as promising anti-liver fibrosis agents.
Collapse
Affiliation(s)
- Jianing Tian
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | - Shicheng Fan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China
| | - Xingrong Peng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Haiguo Su
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen 518055, China.
| | - Minghua Qiu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
104
|
Zhou W, Wang J, Ding L, Cai R, Cheng J, Liang P, Zhu Y, Zhang Z. Autophagy inhibition enhances antifibrotic potential of placental mesenchymal stem cells of fetal origin via regulating TGF-β1 mediated protein degradation of HGF. Sci Rep 2025; 15:13805. [PMID: 40258831 PMCID: PMC12012010 DOI: 10.1038/s41598-025-97054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/02/2025] [Indexed: 04/23/2025] Open
Abstract
Mesenchymal stem cell (MSC) therapy represents a promising strategy for pulmonary fibrosis (PF) treatment, with hepatocyte growth factor (HGF) serving as a key mediator of MSC-mediated protection. However, the therapeutic efficacy of MSCs is limited by the complex PF microenvironment, and the mechanisms underlying this limitation remain unclear. This study investigates how the PF pathological microenvironment modulates the antifibrotic potential of placental mesenchymal stem cells of fetal origin (fPMSCs) through HGF regulation and elucidates the molecular mechanisms involved. Morphological analysis, flow cytometry, and multilineage differentiation assays were employed to characterize fPMSCs. Transforming growth factor-β1 (TGF-β1) was employed to simulate the PF microenvironment and activate fPMSCs in vitro. ELISA and Western blotting were used to analyze HGF expression, autophagy markers, and Smad signaling. Autophagosome formation was visualized via confocal microscopy and transmission electron microscopy. Co-immunoprecipitation (Co-IP) assays were performed to assess the interaction between p62 and HGF. The antifibrotic function of fPMSCs was further evaluated using a transwell co-culture system with MRC-5 fibroblasts in vitro and a bleomycin-induced PF mouse model in vivo. Phenotypic characterization confirmed that fPMSCs exhibited canonical MSC morphology, expressed CD73/CD90/CD105, lacked CD14/CD34/CD45/HLA-DR, and differentiated into adipogenic, osteogenic, and chondrogenic lineages. TGF-β1 treatment robustly downregulated the antifibrotic capacity, HGF protein expression, and paracrine secretion in fPMSCs. Recombinant HGF enhanced antifibrotic effects, while an HGF-neutralizing antibody abolished them. TGF-β1 induced autophagy in fPMSCs, promoting HGF degradation via p62 interaction and impairing antifibrotic function in vitro and in vivo. Mechanistically, Smad3 phosphorylation mediated the regulation of autophagy and HGF expression in TGF-β1-treated fPMSCs. Our findings demonstrate that TGF-β1 impairs the antifibrotic function of fPMSCs via autophagy-dependent HGF degradation and Smad3 signaling. Conversely, autophagy inhibition restores HGF levels and enhances fPMSCs' therapeutic efficacy in a preclinical PF model. Targeting autophagy inhibition emerges as a promising therapeutic strategy to counteract pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei Zhou
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- The People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Jing Wang
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Lu Ding
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, 804 W. Shengli Road, Yinchuan, 750000, Ningxia, China
| | - Ruizhi Cai
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Jie Cheng
- The Third School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Panpan Liang
- The People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750000, Ningxia, China
| | - Yongzhao Zhu
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, 804 W. Shengli Road, Yinchuan, 750000, Ningxia, China.
| | - Zaiqi Zhang
- The First School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750000, Ningxia, China.
| |
Collapse
|
105
|
Oishi K, Yoshida Y, Kaida K, Terai K, Yamamoto H, Toyoda A. Potential non-invasive biomarkers of chronic sleep disorders identified by salivary metabolomic profiling among middle-aged Japanese men. Sci Rep 2025; 15:10980. [PMID: 40258870 PMCID: PMC12012070 DOI: 10.1038/s41598-025-95403-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
Sleep disorders have become a global social problem that increases the risk of developing mental illnesses and metabolic diseases. We aimed to identify biomarkers with which to non-invasively and objectively evaluate chronic sleep disorders. We used capillary electrophoresis-Fourier transform mass spectrometry (CE-FTMS) to analyze metabolomes in saliva collected from 50 persons each with good (≤ 2) and poor (≥ 6) sleep quality scored according to the Japanese version of the Pittsburgh Sleep Quality Index (PSQI-J) self-report questionnaire. The levels of five metabolites including glycerol and hippuric acid and eight including 2-hydroxybutyric acid (2HB), were respectively decreased and increased in participants with poor sleep quality. We established a random forest model consisting of six metabolites, including glycerol and hippuric acid, with a prediction accuracy of 0.866. Correlations between metabolites and sleep satisfaction were assessed using the Oguri-Shirakawa-Azumi sleep inventory, middle-age and aged version (OSA-MA) questionnaire. The results showed that 2'-deoxyguanosine, N1-acetylspermine, and 2,4-dihydroxybenzoic acid correlated positively, whereas glucosamine 6-phosphate and trimethylamine N-oxide correlated negatively with sleep quality. These findings suggested that changes in salivary metabolites reflect pathophysiological mechanisms of chronic sleep disorders, and that saliva samples could serve as non-invasive and objective diagnostic targets for predicting habitual sleep quality.
Collapse
Affiliation(s)
- Katsutaka Oishi
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
- Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Chiba, Japan.
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan.
| | - Yuta Yoshida
- Department of Food and Life Sciences, College of Agriculture, Ibaraki University, Ami, Ibaraki, Japan
| | - Kosuke Kaida
- Institute for Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Kozue Terai
- Human Metabolome Technologies Inc, Tsuruoka, Yamagata, Japan
| | | | - Atsushi Toyoda
- Department of Food and Life Sciences, College of Agriculture, Ibaraki University, Ami, Ibaraki, Japan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| |
Collapse
|
106
|
Zhang Y, Ye L, Qin Y, Qiu C, Sun Q, Fan T, Chen Y, Jiang Y. Serum metabolomics to identify molecular subtypes and predict XELOX efficacy in colorectal cancer. Sci Rep 2025; 15:13671. [PMID: 40258977 PMCID: PMC12012017 DOI: 10.1038/s41598-025-97463-9] [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: 11/25/2024] [Accepted: 04/04/2025] [Indexed: 04/23/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers; however, accurately predicting prognosis based on existing molecular subtypes remains challenging. The XELOX regimen, which combines oxaliplatin and capecitabine, is the cornerstone of chemotherapy for CRC treatment. However, there is a notable lack of reliable predictive models for determining the sensitivity of this treatment. This study aimed to establish a novel classification system for CRC and develop a predictive model for XELOX chemotherapeutic sensitivity using serum metabolomics. We recruited 89 patients with CRC and 89 age- and sex-matched healthy controls for untargeted metabolomic studies to identify tumor-specific serum metabolites. The patients were grouped into distinct metabolic subtypes using unsupervised clustering. A serum metabolite combination predictive of the efficacy of XELOX was established using Cox regression analysis in 34 patients with stage III CRC. Using unsupervised clustering based on the serum metabolites, three distinct clusters were identified. Notably, Cluster 3, which was characterized by uniform lipid and amino acid levels, demonstrated the best prognosis. Our analysis revealed that D-glucose 6-phosphate, presqualene diphosphate, and leukotriene B4 levels were negatively correlated with XELOX sensitivity, whereas 15-HETE and N-acetyl-l-methionine levels were positively correlated. Based on these findings, we constructed a predictive model validated in an independent cohort of 34 patients with stage III CRC. In summary, this study identified a novel classification of CRC based on serum metabolites and developed a potential prognostic model for XELOX chemotherapeutic efficacy, which may have direct effects on the treatment and prognosis of CRC.
Collapse
Affiliation(s)
- Yijie Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Lizhen Ye
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People'S Hospital, Shenzhen, China
| | - Cheng Qiu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Qinsheng Sun
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yan Chen
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China.
| | - Yuyang Jiang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
107
|
Yu P, Liu B, Dong C, Chang Y. Induced Pluripotent Stem Cells-Based Regenerative Therapies in Treating Human Aging-Related Functional Decline and Diseases. Cells 2025; 14:619. [PMID: 40277944 PMCID: PMC12025799 DOI: 10.3390/cells14080619] [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: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
A significant increase in life expectancy worldwide has resulted in a growing aging population, accompanied by a rise in aging-related diseases that pose substantial societal, economic, and medical challenges. This trend has prompted extensive efforts within many scientific and medical communities to develop and enhance therapies aimed at delaying aging processes, mitigating aging-related functional decline, and addressing aging-associated diseases to extend health span. Research in aging biology has focused on unraveling various biochemical and genetic pathways contributing to aging-related changes, including genomic instability, telomere shortening, and cellular senescence. The advent of induced pluripotent stem cells (iPSCs), derived through reprogramming human somatic cells, has revolutionized disease modeling and understanding in humans by addressing the limitations of conventional animal models and primary human cells. iPSCs offer significant advantages over other pluripotent stem cells, such as embryonic stem cells, as they can be obtained without the need for embryo destruction and are not restricted by the availability of healthy donors or patients. These attributes position iPSC technology as a promising avenue for modeling and deciphering mechanisms that underlie aging and associated diseases, as well as for studying drug effects. Moreover, iPSCs exhibit remarkable versatility in differentiating into diverse cell types, making them a promising tool for personalized regenerative therapies aimed at replacing aged or damaged cells with healthy, functional equivalents. This review explores the breadth of research in iPSC-based regenerative therapies and their potential applications in addressing a spectrum of aging-related conditions.
Collapse
Affiliation(s)
- Peijie Yu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Bin Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Cheng Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Yun Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China; (P.Y.); (B.L.)
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
108
|
Yu Y, Yang X, Deng J, Yin Y, Wu Y, Yu R. Association of the gut microbiome with diabetic nephropathy and the mediated effect of metabolites: friend or enemy? Int Urol Nephrol 2025:10.1007/s11255-025-04519-w. [PMID: 40257664 DOI: 10.1007/s11255-025-04519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE The effects of gut microbiome and its metabolites on diabetic nephropathy (DN) have been inadequately elucidated. The aim of this study is to assess the causal effect of gut microbiome on DN and the mediated effect of metabolites by a two-step Mendelian randomization (MR). METHODS Datasets of gut microbiome, metabolites, and DN were acquired in genome-wide association studies and screened for single nucleotide polymorphisms according to the underlying assumptions of MR. Subsequently, inverse variance weighted was used as the primary method for MR analysis to assess the causal effect of gut microbiome on DN and the mediated effect of metabolites. Finally, MR-Egger intercept, Cochran's Q test, and leave-one-out sensitivity analysis were used to assess the horizontal pleiotropy, heterogeneity, and robustness of the results, respectively. RESULTS The MR analysis demonstrated that Parabacteroides merdae increased the genetic susceptibility to DN by reducing acetylcarnitine (C2) to propionylcarnitine (C3) ratio (mediated proportion 8.95%, mediated effect 0.024) and alpha-ketobutyrate to 3-methyl-2-oxovalerate ratio (mediated proportion 19.90%, mediated effect 0.053). MR Egger showed that these results lack horizontal pleiotropy (p ≥ 0.05). Cochran's Q and sensitivity analysis suggested these results had no heterogeneity (p ≥ 0.05) and were robust. CONCLUSION Our findings revealed the pathway by which Parabacteroides merdae increased the genetic susceptibility to DN by regulating acetylcarnitine (C2) to propionylcarnitine (C3) ratio and alpha-ketobutyrate to 3-methyl-2-oxovalerate ratio. It provides new genetic insights for understanding the pathogenesis of DN and related drug research.
Collapse
Affiliation(s)
- Yunfeng Yu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyu Yang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Juan Deng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuman Yin
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yongjun Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| | - Rong Yu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
109
|
Luo X, Yang W, Kang Y, Ma S, Fan Y, Du J, Luo H, Wang X, Deng F. Research progress of the intestinal axis in autologous arteriovenous fistula stenosis in maintenance hemodialysis patients. J Vasc Access 2025:11297298251332047. [PMID: 40251786 DOI: 10.1177/11297298251332047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2025] Open
Abstract
The number of the patients treated with maintenance hemodialysis (HD) is increasing due to the increasing incidence of end stage renal disease (ESRD). Autologous arteriovenous fistula (AVF) is the preferable modality for long-term vascular access during HD. AVF stenosis is the main cause of AVF dysfunction in HD patients, but its mechanism has not been fully elucidated. Patients with ESRD often have various related complications due to intestinal microbiota disorders and their metabolites, and the intestinal axis reveals various metabolic disorders in patients with chronic kidney disease. This paper analyzes the correlation between intestinal axis abnormalities and AVF stenosis in patients with CKD through three axes: "gut-liver axis," "gut-brain axis," and "gut-spleen axis," to provide clinical significance for elucidating the mechanism of AVF stenosis and for the prevention and treatment of AVF stenosis.
Collapse
Affiliation(s)
- Xuyang Luo
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Sichuan Provincial People's Hospital Xinjin Hospital, Chengdu Xinjin District People's Hospital, Chengdu, China
| | - Wei Yang
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuwei Kang
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shijie Ma
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Fan
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiaojiao Du
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Luo
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xudong Wang
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Fei Deng
- Department of Nephrology, Sichuan Provincial People's Hospital Xinjin Hospital, Chengdu Xinjin District People's Hospital, Chengdu, China
| |
Collapse
|
110
|
Ma X, Guan B, Pang L. Calycosin ameliorates albuminuria in nephrotic syndrome by targeting Notch1/Snail pathway. BMC Nephrol 2025; 26:198. [PMID: 40251522 PMCID: PMC12008911 DOI: 10.1186/s12882-025-04113-3] [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: 02/19/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Heavy proteinuria is an important hallmark for kidney disease including nephrotic syndrome. Astragali Radix, a traditional Chinese herb, holds the potential to alleviate nephrotic syndrome; however, the underlying mechanism has not been completely clarified. The study aimed to explore the role of calycosin (C16H12O5), a major active component of Astragali Radix, in regulating adriamycin-induced proteinuria. METHODS A rat model of nephrotic syndrome was established through two adriamycin injections within two weeks (4 mg/kg for the first week and 2 mg/kg for the second week). After the induction of renal injury, 10 mg/kg or 20 mg/kg calycosin was intraperitoneally injected into rats for four weeks. Before euthanasia of rats, urine and blood samples were collected, and body weight was recorded. Then, 24 h urine protein content, kidney index, total cholesterol (TC), triglyceride (TG), as well as renal function indicators including blood urea nitrogen (BUN), serum creatinine (SCR), and urine albumin excretory rate (UAE) were measured. Hematoxylin-eosin staining for renal cortex tissues was performed to evaluate glomerular structural damage. TUNEL assay was performed to evaluate renal cell apoptosis. Western blotting was conducted to measure protein levels of podocyte-specific markers (podocin and nephrin), Notch1, and Snail in rat renal tissues. RESULTS Calycosin reversed adriamycin-induced increase in proteinuria content, kidney index, and concentrations of renal function indicators. Calycosin ameliorated glomerular structural damage, inflammatory cell infiltration, and basement membrane thickening in model rats. In addition, calycosin rescued the suppressive impact of adriamycin on renal cell apoptosis and protein levels of podocyte markers. The activated Notch1/Snail signaling in model rats was suppressed by calycosin intervention. CONCLUSION Calycosin exerts a protective role against adriamycin-induced nephrotic syndrome via inhibition of the Notch1/Snail signaling. CLINICAL TRIAL DETAILS Not applicable.
Collapse
Affiliation(s)
- Xiaohong Ma
- Department of Nephrology, Shenzhen Bao'an Authentic TCM Therapy Hospital, Room 1703, Block G, Jiazhou Business Center, Baomin 1 Road, Xin 'an Street, Bao 'an District, Shenzhen, Guangdong, 518100, China.
| | - Binghe Guan
- Department of Internal Medicine, Shenzhen Bao'an Authentic TCM Therapy Hospital, Shenzhen, 518100, China
| | - Linrong Pang
- Department of Internal Medicine, Shenzhen Bao'an Authentic TCM Therapy Hospital, Shenzhen, 518100, China
| |
Collapse
|
111
|
Adamowicz K, Lima Ribeiro AS, Golda A, Wadowska M, Potempa J, Schmaderer C, Anders HJ, Koziel J, Lech M. Bidirectional Interaction Between Chronic Kidney Disease and Porphyromonas gingivalis Infection Drives Inflammation and Immune Dysfunction. J Immunol Res 2025; 2025:8355738. [PMID: 40276114 PMCID: PMC12021489 DOI: 10.1155/jimr/8355738] [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: 03/15/2024] [Revised: 12/22/2024] [Accepted: 02/04/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction: Chronic kidney disease (CKD) is characterized by a decline in renal function, increased mortality, and significant impairments in the immune system and function of immune cells. These alterations are often derived by uremic toxins, which, in turn, modify the immune system's response to infections. Our research investigates the progression of Porphyromonas gingivalis (P. gingivalis) infection during CKD and its subsequent impact on kidney failure. Methods: We utilized two infectious models, a chamber model representing short-term local inflammation and alveolar bone loss that mimic chronic infection of periodontium, both in conjunction with a CKD model. Additionally, our in vitro studies employed primary macrophages, osteoclasts, and lymphocytes to characterize the immune responses to P. gingivalis and pathogen-associated molecular patterns (PAMPs) in the presence of uremic toxins. Results and Conclusion: Our findings demonstrate that uremic toxins, such as indoxyl sulfate (IS), alter responses of macrophages and lymphocytes to P. gingivalis. In vivo, CKD significantly enhanced P. gingivalis survival and infection-induced alveolar bone loss. The increased distribution of pathogen within peripheral tissues was associated with altered inflammatory responses, indicating that CKD promotes infection. Moreover, P. gingivalis-infected mice exhibited a marked increase in renal inflammation, suggesting that the relationship between uremia and infection is bidirectional, with infection exacerbating kidney dysfunction. Furthermore, we observed that infected CKD mice exhibit decreased serum immunoglobulin G (IgG) levels compared to infected mice without CKD, implying that uremia is associated with immune dysfunction characterized by immunodepression and impaired B lymphocyte function.
Collapse
Affiliation(s)
- Karina Adamowicz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Andrea Sofia Lima Ribeiro
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- TUM University Hospital, Technical University Munich (TUM), Munich, Germany
| | - Anna Golda
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Marta Wadowska
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
- Department of Oral Immunity and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | | | - Hans-Joachim Anders
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Maciej Lech
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
112
|
Adugna A, Amare GA, Jemal M. Machine Learning Approach and Bioinformatics Analysis Discovered Key Genomic Signatures for Hepatitis B Virus-Associated Hepatocyte Remodeling and Hepatocellular Carcinoma. Cancer Inform 2025; 24:11769351251333847. [PMID: 40291818 PMCID: PMC12033511 DOI: 10.1177/11769351251333847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Hepatitis B virus (HBV) causes liver cancer, which is the third most common cause of cancer-related death worldwide. Chronic inflammation via HBV in the host hepatocytes causes hepatocyte remodeling (hepatocyte transformation and immortalization) and hepatocellular carcinoma (HCC). Recognizing cancer stages accurately to optimize early screening and diagnosis is a primary concern in the outlook of HBV-induced hepatocyte remodeling and liver cancer. Genomic signatures play important roles in addressing this issue. Recently, machine learning (ML) models and bioinformatics analysis have become very important in discovering novel genomic signatures for the early diagnosis, treatment, and prognosis of HBV-induced hepatic cell remodeling and HCC. We discuss the recent literature on the ML approach and bioinformatics analysis revealed novel genomic signatures for diagnosing and forecasting HBV-associated hepatocyte remodeling and HCC. Various genomic signatures, including various microRNAs and their associated genes, long noncoding RNAs (lncRNAs), and small nucleolar RNAs (snoRNAs), have been discovered to be involved in the upregulation and downregulation of HBV-HCC. Moreover, these genetic biomarkers also affect different biological processes, such as proliferation, migration, circulation, assault, dissemination, antiapoptosis, mitogenesis, transformation, and angiogenesis in HBV-infected hepatocytes.
Collapse
Affiliation(s)
- Adane Adugna
- Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Ethiopia
| | - Gashaw Azanaw Amare
- Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Ethiopia
| | - Mohammed Jemal
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Ethiopia
| |
Collapse
|
113
|
Cheng X, Hao W, Yu S, Gao X, Qu L, Liu C, Wang Y, Sun Y, Huang J, Yang L, Wang J. Nephroprotective effects of Amomum kravanh essential oil by inhibition of ferroptosis regulated by Nrf2/HO-1 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156762. [PMID: 40305973 DOI: 10.1016/j.phymed.2025.156762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Amomum kravanh Pierre ex Gagnep. (BDK) is a Zingiberaceae plant traditionally widely used as a sweet fragrance, and commonly also utilized in minority medicine for various kidney diseases, especially chronic kidney disease (CKD) in Tibetan and Mongolian medicine. However, the underlying mechanisms by which it confers renal protection remain to be fully clarified. PURPOSE To investigate the renal protective mechanism of which BDK's essential oil exerts in rats with CKD induced by adenine and 5/6 nephrectomy. METHODS Rat models of adenine and 5/6 nephrectomy chronic nephropathy were established, and the therapeutic effects were evaluated by detecting the blood biochemical levels and H&E-/Masson staining and fiber-related factors. Then, the chemical composition of BDK's essential oil and blood components were analyzed using GC-MS. The efficacy of eucalyptol was evaluated by adenine and 5/6 nephrectomy CKD model, with mechanistic studies conducted using RNA-seq, western blot, and metabolomic approaches. RESULTS The blood biochemical levels and histopathological analyses (H&E-/Masson's staining) revealed that the BDK's essential oil significantly enhanced renal function and ameliorated kidney tissue fibrosis. Furthermore, GC-MS analysis identified 33 components in the essential oil of BDK, with eucalyptol being the predominant chemical component at 74.07 %. Eucalyptol is capable of entering the bloodstream in its prototypical form. Then, the efficacy and mechanism of eucalyptol were confirmed by adenine/5/6 nephrectomy CKD models, and based on RNA-seq analysis, we found that eucalyptol could significantly improve kidney function and fibrosis of kidney tissues by blocking TGF-β/smad and NF-κB pathways and inhibit ferroptosis through the Nrf2/HO-1 signaling pathway. CONCLUSION Both BDK's essential oil and its main constituent, eucalyptol, exhibited protective effects against CKD. They both ameliorated oxidative stress, inflammation, and fibrosis in adenine/5/6 nephrectomy rats. Eucalyptol is implicated in ferroptosis and regulation of renal fibrosis via the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Xiaoling Cheng
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Wenli Hao
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Silin Yu
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Laboratory of Fruit Tree Species Breeding and Cultivation in Xinjiang, Urumqi, Xingjiang 830052, China
| | - Xvjie Gao
- School of Pharmacy, Shihezi University/Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, Shihezi, Xinjiang, 832002, China
| | - Liyuan Qu
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chang Liu
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yanli Wang
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yifan Sun
- Shenzhen Honghui Biopharmaceutical Co., Ltd. Shenzhen 518000, China
| | - Jian Huang
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| | - Lu Yang
- Key Laboratory of Fruit Tree Species Breeding and Cultivation in Xinjiang, Urumqi, Xingjiang 830052, China.
| | - Jinhui Wang
- School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
114
|
Zhao Q, Jin M, Zhao Q, Wang Z, Zhao C, Xue X, Qiao X, Qu P, Han D, Tao R. Natural products in traditional Chinese medicine for renal fibrosis: a comprehensive review. Front Pharmacol 2025; 16:1560567. [PMID: 40308781 PMCID: PMC12041090 DOI: 10.3389/fphar.2025.1560567] [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: 01/14/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Renal fibrosis represents the terminal pathological manifestation of most chronic kidney diseases, driving progressive loss of renal function. Natural products have emerged as promising therapeutic agents for preventing and ameliorating renal fibrosis due to their multi-target efficacy and favorable safety profiles. In this review, we conducted a comprehensive literature search on PubMed using the keywords "natural product" and "renal fibrosis" from 2004 to 2025, identifying 704 relevant articles. We systematically categorize and discuss the biological effects of key natural products and formulations with antifibrotic potential, focusing on five major classes: glycosides, flavonoids, phenolic compounds, anthraquinones, and terpenoids. Representative compounds from each category are highlighted for their mechanisms of action, including modulation of oxidative stress, inflammation, autophagy, and fibrosis signaling pathways. This review aims to provide a theoretical foundation for the development of natural product-based therapies to combat renal fibrosis, offering insights into their therapeutic potential and future research directions.
Collapse
Affiliation(s)
- Qianqian Zhao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Meihua Jin
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Qiang Zhao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Zhimei Wang
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Chun Zhao
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xiaocong Xue
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xikai Qiao
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Peng Qu
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Donghe Han
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| |
Collapse
|
115
|
Cao H, Li Z, Ye J, Lv Y, Zhang C, Liang T, Wang Y. Emerging roles of exosomes in the diagnosis and treatment of kidney diseases. Front Pharmacol 2025; 16:1525314. [PMID: 40308771 PMCID: PMC12041035 DOI: 10.3389/fphar.2025.1525314] [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: 11/09/2024] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
The complex etiology and spectrum of kidney diseases necessitate vigilant attention; the focus on early diagnosis and intervention in kidney diseases remains a critical issue in medical research. Recently, with the expanding studies on extracellular vesicles, exosomes have garnered increasing interest as a promising tool for the diagnosis and treatment of kidney diseases. Exosomes are nano-sized extracellular vesicles that transport a diverse array of bioactive substances, which can influence various pathological processes associated with kidney diseases and exhibit detrimental or beneficial effects. Within the kidney, exosomes derived from the glomeruli and renal tubules possess the ability to enter systemic circulation or urine. The biomarkers they carry can reflect alterations in the pathological state of the kidneys, thereby offering novel avenues for early diagnosis. Furthermore, research studies have confirmed that exosomes originating from multiple cell types exhibit therapeutic potential in treating kidney disease; notably, those derived from mesenchymal stem cells (MSCs) have shown significant treatment efficacy. This comprehensive review summarizes the contributions of exosomes from different cell types within the kidneys while exploring their physiological and pathological roles therein. Additionally, we emphasize recent advancements in exosome applications for the diagnosis and treatment of various forms of kidney diseases over the past decades. We not only introduce the urinary and blood biomarkers linked to kidney diseases found within exosomes but also explore their therapeutic effects. Finally, we discuss existing challenges and future directions concerning the clinical applications of exosomes for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Huanhuan Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixi Li
- Department of Clinical Laboratory, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajia Ye
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Lv
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Liang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yumei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
116
|
Wang D, Hou P, Lang H, Xia Y, Bai Q, Yao Y, Yi L, Mi M. L-Tryptophan-Rich Diet Alleviates High-Intensity-Exercise-Induced Liver Dysfunction via the Metabolite Indole-3-Acetic Acid and AhR Activation. Cells 2025; 14:605. [PMID: 40277929 PMCID: PMC12026455 DOI: 10.3390/cells14080605] [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: 03/16/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
High-intensity exercise (HIE) induces liver dysfunction and is detrimental to exercise performance. The underlying mechanism and preventive strategy urgently need to be explored. We increased the amount of tryptophan appropriately in the diet and explored the effect of an L-tryptophan-rich diet on the alleviation of HIE-induced liver dysfunction and the underlying mechanism. In this work, by establishing a C57BL/6 mouse model of high-intensity swimming exercise, the results demonstrated an L-tryptophan-rich diet significantly attenuated HIE-induced liver dysfunction, which was associated with increased levels of the tryptophan metabolite indole-3-acetic acid (IAA). Furthermore, IAA indeed exerted a protective effect against HIE-induced liver dysfunction in vivo and LPS-induced hepatocyte dysfunction in vitro. In conclusion, an L-tryptophan-rich diet may be a promising strategy to prevent HIE-induced liver dysfunction and metabolic disturbance via the metabolite indole-3-acetic acid and AhR activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mantian Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Health, Chongqing Medical Nutrition Research Center, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing 400038, China; (D.W.); (P.H.); (H.L.); (Y.X.); (Q.B.); (Y.Y.); (L.Y.)
| |
Collapse
|
117
|
Cho Y, Seo CW, Cho H, Jin Y, Lupala AS, Shim SH, Lim YW. A conserved terpene cyclase gene in Sanghuangporus for abscisic acid-related sesquiterpenoid biosynthesis. BMC Genomics 2025; 26:378. [PMID: 40234762 PMCID: PMC12001456 DOI: 10.1186/s12864-025-11542-9] [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: 10/09/2024] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND The medicinal mushroom Sanghuangporus is renowned in East Asia for its potent therapeutic properties, attributed in part to its bioactive sesquiterpenoids. However, despite their recognized medicinal potential, the biosynthetic pathways and specific enzymes responsible for sesquiterpenoid production in Sanghuangporus remain unexplored, limiting opportunities to optimize their medicinal applications. RESULTS Sesquiterpenoids from four Sanghuangporus species were extracted through targeted isolation using mass spectrometry (MS)-based metabolomics, resulting in the discovery of six known abscisic acid-related compounds and one new compound, whose structure was determined through spectroscopic and computational analysis. We employed a natural product genome mining approach to identify a putative biosynthetic gene cluster (BGC) containing a sesquiterpene synthase gene, ancA, associated with the detected compounds. Biosynthetic pathways for these compounds were proposed based on an integrative approach combining BGC analysis and MS2 fragment-based dereplication. Further analyses revealed that the gene content and synteny of the ancA BGC are relatively well-conserved across Sanghuangporus species but less so outside the genus. CONCLUSIONS A sesquiterpene synthase gene, its associated BGC, and the biosynthetic pathway for a group of detected abscisic acid-related sesquiterpenoids in Sanghuangporus were predicted through genomic and metabolic data analyses. This study addresses a critical gap in understanding the genetic basis of sesquiterpenoid production in Sanghuangporus and offers insights for future research on engineering metabolic pathways to enhance sesquiterpenoid production for medicinal use.
Collapse
Affiliation(s)
- Yoonhee Cho
- School of Biological Sciences and Institute of Biodiversity, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chang Wan Seo
- School of Biological Sciences and Institute of Biodiversity, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyeonjae Cho
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeongwoon Jin
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Abel Severin Lupala
- School of Biological Sciences and Institute of Biodiversity, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Microbiology, Parasitology and Biotechnology, Sokoine University of Agriculture, P.O. Box 3019, Morogoro, 67125, Tanzania
| | - Sang Hee Shim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Young Woon Lim
- School of Biological Sciences and Institute of Biodiversity, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
118
|
Bosetti C, Kampasis D, Brinch SA, Galera-Prat A, Karelou M, Dhakar SS, Alaviuhkola J, Waaler J, Lehtiö L, Kostakis IK. Substitutions at the C-8 position of quinazolin-4-ones improve the potency of nicotinamide site binding tankyrase inhibitors. Eur J Med Chem 2025; 288:117397. [PMID: 39983556 DOI: 10.1016/j.ejmech.2025.117397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Human diphtheria toxin-like ADP-ribosyltransferases, PARPs and tankyrases, transfer ADP-ribosyl groups to other macromolecules, thereby controlling various signaling events in cells. They are considered promising drug targets, especially in oncology, and a vast number of inhibitors have already been successfully developed. These inhibitors typically occupy the nicotinamide binding site and extend along the NAD+ binding groove of the catalytic domain. Quinazolin-4-ones have been explored as compelling scaffolds for such inhibitors and we have identified a new position within the catalytic domain that has not been extensively studied yet. In this study, we investigate larger substituents at the C-8 position and, using X-ray crystallography, we demonstrate that nitro- and diol-substituents engage in new interactions with TNKS2, improving both affinity and selectivity. Both diol- and nitro-substituents exhibit intriguing inhibition of TNKS2, with the diol-based compound EXQ-1e displaying a pIC50 of 7.19, while the nitro-based compound EXQ-2d's pIC50 value is 7.86. Both analogues impact and attenuate the tankyrase-controlled WNT/β-catenin signaling with sub-micromolar IC50. When tested against a wider panel of enzymes, the nitro-based compound EXQ-2d displayed high selectivity towards tankyrases, whereas the diol-based compound EXQ-1e also inhibited other PARPs. Compound EXQ-2d displays in vitro cell growth inhibition of the colon cancer cell line COLO 320DM, while compound EXQ-1e displays nonspecific cell toxicity. Collectively, the results offer new insights for inhibitor development targeting tankyrases and PARPs by focusing on the subsite between a mobile active site loop and the canonical nicotinamide binding site.
Collapse
Affiliation(s)
- Chiara Bosetti
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Dionysis Kampasis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Shoshy A Brinch
- Oslo University Hospital, P.O. Box 4950, Nydalen, Oslo, 0424, Norway; Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Albert Galera-Prat
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Maria Karelou
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Saurabh S Dhakar
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Juho Alaviuhkola
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Jo Waaler
- Oslo University Hospital, P.O. Box 4950, Nydalen, Oslo, 0424, Norway; Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland.
| | - Ioannis K Kostakis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, 15771, Athens, Greece.
| |
Collapse
|
119
|
Chen Q, Song Y, Wu Q, Wu Y, Zhou M, Ren Y, Guo X, Cao G, Li B, Duan Z, Gao P. Dietary Angelica sinensis Enhances Sow Lactation and Piglet Development Through Gut Microbiota and Metabolism. Vet Sci 2025; 12:370. [PMID: 40284872 PMCID: PMC12030784 DOI: 10.3390/vetsci12040370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Piglets weaned per sow per year (PSY) is a crucial metric for assessing the reproductive performances of sows and directly affects both sow productivity and the economic profitability of pig farms [...].
Collapse
Affiliation(s)
- Qian Chen
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| | - Yali Song
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| | - Qitian Wu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| | - Yali Wu
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China; (Y.W.); (M.Z.)
| | - Maocuo Zhou
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China; (Y.W.); (M.Z.)
| | - Yifei Ren
- Tiankang Livestock Technology Company Limited, Zhumadian 463343, China;
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| | - Zhibian Duan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China;
| | - Pengfei Gao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (Q.C.); (Y.S.); (Q.W.); (X.G.); (G.C.); (B.L.)
| |
Collapse
|
120
|
Gorman BL, Lukowski JK. Spatial Metabolomics and Lipidomics in Kidney Disease. Semin Nephrol 2025:151582. [PMID: 40234137 DOI: 10.1016/j.semnephrol.2025.151582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Kidney disease is a global health issue that affects over 850 million people, and early detection is key to preventing severe disease and complications. Kidney diseases are associated with complex and dysregulation of lipid metabolism. Spatial metabolomics through mass spectrometry imaging (MSI) enables spatial mapping of the lipids in tissue and includes a variety of techniques that can be used to image lipids. In the kidney, MSI studies often seek to resolve individual functional tissue units such as glomeruli and proximal tubules. Several different MSI techniques, such as matrix-assisted laser desorption/ionization (MALDI) and desorption electrospray ionization (DESI), have been used to characterize lipids and small molecules in chronic kidney disease, acute kidney injury, genetic kidney disease, and cancer. In this review we provide several examples of how spatial metabolomics data can provide critical information concerning the localization of changes in various disease states. Additionally, when combined with pathology, transcriptomics, or proteomics, the metabolomic changes can illuminate underlying mechanisms and provide new clinical insights into disease mechanisms. Semin Nephrol 36:x-xx © 20xx Elsevier Inc. All rights reserved.
Collapse
Affiliation(s)
| | - Jessica K Lukowski
- Mass Spectrometry Imaging Lead, Mass Spectrometry Technology Access Center at the McDonnell Genome Institute, Washington University in St. Louis School of Medicine, St. Louis, MO
| |
Collapse
|
121
|
Huang R, Che R, Sun T, Xie W, Zhang S. Investigating the renoprotective effects of Polygonatum sibiricum polysaccharides (PSP) on D-galactose-induced aging mice: insights from gut microbiota and metabolomics analyses. Front Microbiol 2025; 16:1550971. [PMID: 40303470 PMCID: PMC12037495 DOI: 10.3389/fmicb.2025.1550971] [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: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Polygonatum sibiricum polysaccharides (PSP) have been suggested to possess various health benefits, including anti-aging and renoprotective effects. However, the mechanisms underlying PSP's protective effects on kidney function, particularly in the context of aging, remain unclear. This study explores how PSP protects against D-galactose (D-gal)-induced kidney damage in aging mice, focusing on gut microbiota and metabolomics. Methods Mice were assigned to five groups: control, model (D-gal), vitamin C, low-dose PSP, and high-dose PSP, and treated for 8 weeks. Kidney pathology was assessed via H&E and Masson's trichrome staining. 16S rRNA sequencing analyzed gut microbiota, and non-targeted metabolomics identified metabolic changes. Correlations between gut bacteria and metabolites were examined. Results PSP alleviated renal damage, reducing tubular atrophy, epithelial swelling, and collagen deposition. It increased beneficial gut bacteria (e.g., Lactobacillus, Bifidobacterium) and altered 23 metabolites linked to pathways such as amino acid and sphingolipid metabolism. Gut microbiota and metabolites were strongly correlated, indicating PSP's role in regulating the gut-kidney axis. Conclusion PSP protects against age-related kidney damage by modulating gut microbiota and metabolic pathways, highlighting its therapeutic potential for kidney aging through the gut-kidney axis.
Collapse
Affiliation(s)
- Rui Huang
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Runli Che
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Taoli Sun
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Wen Xie
- The First People's Hospital of Changde City, Changde, China
| | - Shuihan Zhang
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
122
|
Li S, Yang H, Zhang B, Li L, Li X. 5-methoxytryptophan ameliorates renal ischemia/reperfusion injury by alleviating endoplasmic reticulum stress-mediated apoptosis through the Nrf2/HO-1 pathway. Front Pharmacol 2025; 16:1506482. [PMID: 40297140 PMCID: PMC12034636 DOI: 10.3389/fphar.2025.1506482] [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: 10/05/2024] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Background Renal ischemia/reperfusion (I/R) injury is a prevalent clinical complication characterized by high incidence and mortality rates. The endogenous metabolite, 5-Methoxytryptophan (5-MTP), derived from tryptophan, possesses anti-inflammatory and antioxidant properties. However, its role in renal I/R injury remains unclear. In this study, we investigated whether 5-MTP could protect the kidney from I/R injury by ameliorating endoplasmic reticulum stress (ERS)-mediated apoptosis through the Nrf2/HO-1 pathway. Methods and results We established models to examine renal I/R injury in C57BL/6J mice with bilateral renal pedicles clamped and HK-2 cells subjected to hypoxia/reoxygenation (H/R). The administration of 5-MTP improved renal tissue damage and kidney dysfunction impairment and reduced inflammation and oxidative stress. Moreover, 5-MTP attenuated ERS and ERS-mediated apoptosis, while upregulating Nrf2 and HO-1 expression. Additionally, Nrf2-deficient mice and cells were used to determine whether the Nrf2/HO-1 pathway was involved in the role of 5-MTP in alleviating ERS-mediated apoptosis. Nrf2 deficiency led to a partial reduction in the suppressive effects of 5-MTP on inflammation, oxidative stress, and ERS-mediated apoptosis. Conclusion Our findings suggest that 5-MTP alleviates renal I/R injury by inhibiting ERS-related apoptosis via the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Shaona Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongjuan Yang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Zhang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingyu Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangkun Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
123
|
Zhang H, Zhao Z, Liu P, Wang M, Liu YE, He H, Ge Y, Zhou T, Xiao C, You Z, Zhang J. Gastrodin enhances stress resilience through promoting Wnt/β-Catenin-dependent neurogenesis. J Adv Res 2025:S2090-1232(25)00261-9. [PMID: 40233892 DOI: 10.1016/j.jare.2025.04.017] [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: 12/30/2024] [Revised: 03/24/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Enhancing stress resilience constitutes a pivotal strategy in mitigating the risk of depression, making it a critical component of both prevention and treatment. In the current work, we identified a compound, gastrodin (GAS), as capable of enhancing stress resilience, as demonstrated by its ability to protect against depression following chronic stress exposure. OBJECTIVES To elucidate the potential of GAS to promote neurogenesis under chronic stress, along with the associated cellular and molecular processes involved. METHOD We evaluated the effect of GAS on NSPC proliferation and differentiation using both in vitro and in vivo investigations. Neurogenesis was inhibited using temozolomide to verify GAS's impact on stress resilience. Comprehensive methodologies, including hippocampal transcriptome analysis and western blotting, were utilized to identify the involvement of the Wnt/β-catenin pathway. Immunolocalization was conducted to confirm β-catenin's nuclear translocation in SOX2+ cells within the hippocampal dentate gyrus subgranular zone. RESULTS GAS demonstrated robust stimulation of NSPC proliferation and neuronal differentiation, enhancing adult hippocampal neurogenesis under conditions of chronic stress. Inhibition of neurogenesis negated GAS's protective effects on stress resilience. Integrated analysis pointed to the Wnt/β-catenin signaling pathway within NSPCs as a crucial mechanism facilitating GAS-promoted neurogenesis. Inhibiting Wnt expression or blocking β-catenin's nuclear translocation abolished GAS's neurogenic and stress-resilience enhancing effects. CONCLUSION These results suggested that GAS directly activates the Wnt/β-catenin signaling pathway, which promotes the proliferation and neuronal differentiation of NSPCs, thereby enhancing adult hippocampal neurogenesis and promoting stress resilience to mitigate the risk of depression.
Collapse
Affiliation(s)
- Haili Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Zhihuang Zhao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Pei Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Meidan Wang
- Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.
| | - Yu-E Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Hui He
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 6100544, China.
| | - Yangyan Ge
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Zili You
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 6100544, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
124
|
Wan L, Li L, Zhang X, Li B, Brao KJ, Harro JM, Shirtliff ME. Evaluating palm olein and palm stearin on hepatic lipids in C57BL/6 J mice under low and high fat intakes: A quantitative lipidomic approach. Food Chem 2025; 484:144326. [PMID: 40252450 DOI: 10.1016/j.foodchem.2025.144326] [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: 11/30/2024] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
This work aims to study the effects of dietary lipid content and composition on hepatic lipids of mice fed palm olein (PO)-based low-fat diet, PO-based high-fat diet, palm stearin (PS)-based low-fat diet, or PS-based high-fat diet for 8 weeks. Results showed that high PO and PS intake significantly elevated serum LDL-c and TCHO levels and hepatic TG content without altering the expression levels of genes involved in DNL. Overall, quantitative lipidomics revealed that accumulated lipids were polyunsaturated PE and saturated & monounsaturated SM/Cer_NS in response to high PO intake. While high PS intake led to increased levels of polyunsaturated PC and mono- & polyunsaturated TG. These enriched lipids may lead to disturbed hepatic PUFA metabolism. When lipid intake was constant, only under high fat intake did PS show minor effects on hepatic lipids. These results highlighted the role of lipid quantity instead of lipid quality on hepatic lipid profiles.
Collapse
Affiliation(s)
- Liting Wan
- School of Food Science and Technology, Guangdong Provincial Key Laboratory of Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou 510640, China; Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA
| | - Lin Li
- School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, China
| | - Xia Zhang
- School of Food Science and Technology, Guangdong Provincial Key Laboratory of Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Bing Li
- School of Food Science and Technology, Guangdong Provincial Key Laboratory of Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Plant Protein Deep Processing, Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| | - Kristen J Brao
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA
| | - Janette M Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA.
| | - Mark E Shirtliff
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland - Baltimore, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, School of Medicine, University of Maryland - Baltimore, Baltimore, MD 21201, USA
| |
Collapse
|
125
|
He J, Yue H, Zhang S, Dong R, Zhang F, Wang X, Wang K, Zhang H, Yang D, Dong Z, Liu H. Dehydrocorydaline attenuates bleomycin-induced pulmonary fibrosis by inhibiting fibroblast activation. Respir Res 2025; 26:136. [PMID: 40221718 PMCID: PMC11992754 DOI: 10.1186/s12931-025-03218-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is an irreversible, progressive, chronic and fatal interstitial lung disease with limited therapeutic options. Dehydrocorydaline (DHC), derived from the traditional Chinese medicinal plant Corydalis yanhusuo, has exhibited a variety of pharmacological properties. Nevertheless, the potential function and mechanism of DHC in the management of PF have yet to be elucidated. PURPOSE To evaluate the therapeutical efficacy of DHC in different PF models and elucidate its underlying mechanism. METHODS A well-established Bleomycin-induced PF mouse model and human precision-cut lung slices (hPCLS) following fibrosis-inducing cocktail stimulation were employed. The antifibrotic effects of DHC on PF were measured by histopathological manifestation, immunofluorescent staining and expression levels of fibrosis related markers. Human primary pulmonary fibroblasts (HPFs) were used to explore the impact of DHC on fibroblast function and the underlying mechanism. RESULTS Here, we demonstrated that DHC exhibited a therapeutic efficacy in Bleomycin-induced PF mouse model with a dose dependent, as well as in hPCLS after fibrosis-inducing cocktail stimulation, as evidenced by histopathological staining, decrease of Fibronectin, Collagen 1 and α-SMA expression. Additionally, in vitro experiments indicated that DHC effectively suppressed fibroblast to myofibroblast transition, but had no significant effect on the proliferation and migration of fibroblast. Mechanistic studies revealed that the inhibitory effect of DHC on fibroblast activation was dependent on the endoplasmic reticulum stress, thereby inhibiting TGF-β/SMAD signal pathway. CONCLUSIONS Our study implied that DHC hold a promise therapeutic approach against PF by suppressing fibroblast activation. The safety and efficacy of DHC have been preliminary demonstrated in a mouse model.
Collapse
Affiliation(s)
- Jianhan He
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Huihui Yue
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Shufei Zhang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruihan Dong
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Fengqin Zhang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Xuewen Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Huilan Zhang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China
| | - Danlei Yang
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China.
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, No. 41, Northwestern Street, Ningbo, 315010, China.
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Ave, Wuhan, 430030, Hubei, China.
| |
Collapse
|
126
|
Khalili-Tanha G, Radisky ES, Radisky DC, Shoari A. Matrix metalloproteinase-driven epithelial-mesenchymal transition: implications in health and disease. J Transl Med 2025; 23:436. [PMID: 40217300 PMCID: PMC11992850 DOI: 10.1186/s12967-025-06447-w] [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: 02/15/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process in which epithelial cells, defined by apical-basal polarity and tight intercellular junctions, acquire migratory and invasive properties characteristic of mesenchymal cells. Under normal conditions, EMT directs essential morphogenetic events in embryogenesis and supports tissue repair. When dysregulated, EMT contributes to pathological processes such as organ fibrosis, chronic inflammation, and cancer progression and metastasis. Matrix metalloproteinases (MMPs)-a family of zinc-dependent proteases that degrade structural components of the extracellular matrix-sit at the nexus of this transition by dismantling basement membranes, activating pro-EMT signaling pathways, and cleaving adhesion molecules. When normally regulated, MMPs promote balanced ECM turnover and support the cyclical remodeling necessary for proper development, wound healing, and tissue homeostasis. When abnormally regulated, MMPs drive excessive ECM turnover, thereby promoting EMT-related pathologies, including tumor progression and fibrotic disease. This review provides an integrated overview of the molecular mechanisms by which MMPs both initiate and sustain EMT under physiological and disease conditions. It discusses how MMPs can potentiate EMT through TGF-β and Wnt/β-catenin signaling, disrupt cell-cell junction proteins, and potentiate the action of hypoxia-inducible factors in the tumor microenvironment. It discusses how these pathologic processes remodel tissues during fibrosis, and fuel cancer cell invasion, metastasis, and resistance to therapy. Finally, the review explores emerging therapeutic strategies that selectively target MMPs and EMT, ranging from CRISPR/Cas-mediated interventions to engineered tissue inhibitors of metalloproteinases (TIMPs), and demonstrates how such approaches may suppress pathological EMT without compromising its indispensable roles in normal biology.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
127
|
Lei H, Jiang Y, Chen Z, Yao J, Ma W, Huang Y, Zhang P, Xie Z, Zhu L, Tang W. Unveiling the influence of lipidomes on inflammatory bowel disease: a bidirectional mendelian randomization study. BMC Gastroenterol 2025; 25:247. [PMID: 40217472 PMCID: PMC11992711 DOI: 10.1186/s12876-025-03858-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Plasma lipid homeostasis is pivotal in maintaining intestinal health. Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD) as distinct subtypes, manifests unique metabolic signatures. However, the specific roles of lipids in the pathogenesis and therapeutic targeting of IBD remain inadequately explored. This study aims to delineate the genetic influences of plasma lipids on IBD risk. METHODS We obtained genome-wide association study (GWAS) summary statistics of lipidomes and IBD (including UC and CD) from published studies to perform two-sample Mendelian randomization (MR) analyses. Outliers were removed using radial MR, followed by the application of the inverse-variance weighted (IVW) method to assess causal relationships. Sensitivity analyses were also conducted to validate the robustness of the primary results of the MR analyses. Additionally, reverse MR analyses were performed to evaluate the potential for reverse causality. RESULTS The MR analysis identified fourteen lipid species significantly associated with IBD, four with UC, and ten with CD. Phosphatidylcholine (PC; P < 0 .05) and lysophosphatidylcholine (OR = 0.83, P < 0.001) were instrumental in UC, while in CD, alongside these, cholesterol ester (OR = 0.86, P < 0.001), diacylglycerol (OR = 1.21, P = 0.004), and lysophosphatidylethanolamine (OR = 1.30, P < 0.001) also demonstrated causal links. Reverse MR analysis revealed no significant associations between IBDs and 179 lipid species. CONCLUSION This bidirectional MR study has uncovered genetic evidence of a causal relationship between lipidome and IBD, identifying potential therapeutic targets for IBD treatment. The findings suggest that elevated partial phosphatidylcholine, lysophosphatidylcholine, and cholesterol ester levels could reduce the risk of IBD, indicating a potential protective role for these lipid molecules. This study also underscores the critical role of lipidome variability in advancing our understanding of IBD's pathogenic processes and in developing targeted therapies.
Collapse
Affiliation(s)
- Hang Lei
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuhong Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhe Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiaqi Yao
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenjun Ma
- Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Yubei District, Chongqing, 401147, China
| | - Yiqi Huang
- Department of Nephrology, Shaoxing Second Hospital, Shaoxing, 312000, Zhejiang, China
| | - Pengcheng Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhijun Xie
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lv Zhu
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenfu Tang
- Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
128
|
Lee SY, Han K, Kwon HS, Koh ES, Chung S. Fecal Calprotectin as a Prognostic Biomarker for Mortality and Renal Outcomes in Chronic Kidney Disease. Biomolecules 2025; 15:557. [PMID: 40305332 PMCID: PMC12025133 DOI: 10.3390/biom15040557] [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: 03/06/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND/OBJECTIVES Fecal calprotectin (FC) is a biomarker of intestinal inflammation widely used in the assessment of gastrointestinal disorders. However, its role in chronic kidney disease (CKD) remains unclear. Given the growing recognition of the gut-kidney axis in CKD pathophysiology, this study aimed to investigate the association between FC levels, systemic inflammation, renal outcomes, and mortality in CKD patients. METHODS We enrolled a total of 515 CKD patients who underwent fecal calprotectin measurement between 2016 and 2023. After applying the exclusion criteria (inflammatory bowel disease, ongoing renal replacement therapy, or incomplete laboratory data), 260 patients were included in the final analysis and stratified into low-FC (<102 μg/g, n = 130) and high-FC (≥102 μg/g, n = 130) groups based on the median FC value. Factors associated with kidney disease progression and patient survival were analyzed. RESULTS Patients in the high-FC group (≥102 μg/g) were significantly older (72.8 ± 14.63 vs. 64.02 ± 18.15 years, p < 0.0001) and had a higher prevalence of diabetes mellitus (55.38% vs. 42.31%, p = 0.0349), heart failure (21.54% vs. 7.69%, p = 0.0016), and history of acute kidney injury (33.85% vs. 18.46%, p = 0.0048). Elevated FC was independently associated with increased mortality risk (hazards ratio [HR] 1.658, 95% confidence interval [CI] 1.034-2.658, p = 0.0357) with higher mortality rates (48.36 vs. 18.46 per 100,000 person-years). Subgroup analyses revealed stronger associations between FC and mortality in males (HR 2.160, 95% CI 1.046-4.463, p = 0.0375), elderly patients (≥75 years) (HR 2.122, 95% CI 1.209-3.725, p = 0.0088), and non-diabetic patients (HR 2.487, 95% CI 1.141-5.421, p = 0.0219). While FC was not significantly associated with end-stage kidney disease (ESKD) progression (odds ratio [OR] 1.289, 95% CI 0.455-3.650, p = 0.6323), higher FC levels paradoxically predicted slower estimated glomerular filtration rate (eGFR) decline (OR 2.763, 95% CI 1.139-6.699, p = 0.0245). Combined analysis revealed patients with both elevated FC and high-sensitivity C-reactive protein (hs-CRP) had the highest mortality risk (HR 3.504, 95% CI 1.163-10.554, p < 0.0001) compared to those with low levels of both markers. CONCLUSIONS FC is a potential prognostic biomarker for mortality in CKD patients, independently of traditional inflammatory markers. Further research is warranted to elucidate the mechanisms underlying its paradoxical relationship with renal outcomes and its potential role in risk stratification and therapeutic targeting in CKD.
Collapse
Affiliation(s)
- So Young Lee
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea;
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Republic of Korea;
| | - Hyuk-Sang Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea;
| | - Eun Sil Koh
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea;
| | - Sungjin Chung
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea;
| |
Collapse
|
129
|
Kananivand M, Nouri F, Yousefi MH, Pajouhi A, Ghorbani H, Afkhami H, Razavi ZS. Mesenchymal stem cells and their exosomes: a novel approach to skin regeneration via signaling pathways activation. J Mol Histol 2025; 56:132. [PMID: 40208456 DOI: 10.1007/s10735-025-10394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Accelerating wound healing is a crucial objective in surgical and regenerative medicine. The wound healing process involves three key stages: inflammation, cell proliferation, and tissue repair. Mesenchymal stem cells (MSCs) have demonstrated significant therapeutic potential in promoting tissue regeneration, particularly by enhancing epidermal cell migration and proliferation. However, the precise molecular mechanisms underlying MSC-mediated wound healing remain unclear. This review highlights the pivotal role of MSCs and their exosomes in wound repair, with a specific focus on critical signaling pathways, including PI3K/Akt, WNT/β-catenin, Notch, and MAPK. These pathways regulate essential cellular processes such as proliferation, differentiation, and angiogenesis. Moreover, in vitro and in vivo studies reveal that MSCs accelerate wound closure, enhance collagen deposition, and modulate immune responses, contributing to improved tissue regeneration. Understanding these mechanisms provides valuable insights into MSC-based therapeutic strategies for enhancing wound healing.
Collapse
Affiliation(s)
- Maryam Kananivand
- Medical Department, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Nouri
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University (SRBIAU), Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Ali Pajouhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hakimeah Ghorbani
- Department of Sciences, Faculty of Biological Sciences, Tabriz University of Sciences, Tabriz, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran.
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
130
|
Liu B, Zeng M, Kang W, Li S, Wang X, Yu H, Wu H. Integrating serum pharmacochemistry, network pharmacology, and metabolomics to elucidate the detoxification and effect-adjusting mechanism of Chebulae Fructus-processing on Mongolian medicine Euphorbia pekinensis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119582. [PMID: 40054641 DOI: 10.1016/j.jep.2025.119582] [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: 12/20/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia pekinensis Radix (EP) is a traditional medicinal plant widely used in Mongolian and Chinese medicine for its potent therapeutic properties in treating edema, ascites, and various inflammatory conditions. However, EP has toxicity, which can cause swelling and congestion of the gastrointestinal mucosa. Chebulae Fructus is a unique processing method in Mongolian medicine that is believed to mitigate EP's toxicity and adjust its effect, though the mechanisms underlying this detoxification remain poorly understood. AIM OF THE STUDY This study employed an integrative approach combining network pharmacology, serum pharmacochemistry, pharmacology, and metabolomics to investigate the intestinal detoxification and synergistic effects of Chebulae Fructus-processed Euphorbia pekinensis (PEP). MATERIALS AND METHODS The blood-absorbed components of EP and PEP were identified by UPLC-MS/MS. To evaluate holistic effect of the two medicine, network pharmacology was applied to focus on serum components and identify the key compounds and targets mediating effect in addressing five TCM syndromes, as well as modern medicine symptoms including seven types of cancer and myocardial infarction. In terms of detoxification, non-targeted metabolomics was utilized to analyze significant intestinal metabolites and pathways affected by EP and PEP in normal mice. RESULTS A total of 77 and 109 blood-absorbed components were identified from EP and PEP, respectively, including terpenoids, phenolic acids, alkaloids, flavonoids, and tannins. In network pharmacology analysis, key hub genes were identified as therapeutic targets, with PEP exhibiting possibly enhanced effects than EP since it was associated with more targets and diseases in the network. Furthermore, PEP modulated histamine metabolism and arginine biosynthesis pathways, thereby reducing intestinal inflammation. CONCLUSION This study highlights the different anti-cancer and ascites-reducing potential of EP and PEP, emphasizing the detoxification benefits of Chebulae Fructus processing. These findings provide a foundation for the safe and effective therapeutic use of PEP for treating ascites and cancer, while minimizing intestinal toxicity, thereby promoting the safe utilization of medicinal Euphorbiaceae plants.
Collapse
Affiliation(s)
- Bingbing Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; National Base of State Ministry of Education for Inheritance of Chinese Medicine Processing Technology, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Key Laboratory of State Administration of TCM for Standardization of Chinese Medicine Processing, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Jiangsu Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China.
| | - Min Zeng
- College of Pharmacy, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China
| | - Wenqingqing Kang
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR, 999078, China
| | - Shu Li
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR, 999078, China
| | - Xinzhi Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China
| | - Hongli Yu
- College of Pharmacy, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; National Base of State Ministry of Education for Inheritance of Chinese Medicine Processing Technology, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Key Laboratory of State Administration of TCM for Standardization of Chinese Medicine Processing, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Jiangsu Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China.
| | - Hao Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; National Base of State Ministry of Education for Inheritance of Chinese Medicine Processing Technology, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Key Laboratory of State Administration of TCM for Standardization of Chinese Medicine Processing, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China; Jiangsu Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Qixia District, Xianlin Road No. 138, Nanjing, 210023, China.
| |
Collapse
|
131
|
Gao S, Wang X, Xu Q, Li R, Yao L, Zhang A, Zhou Q, Xiao Z, Li S, Meng X, Wu J, Qin L. Total Sanghuangporus vaninii extract inhibits hepatocyte ferroptosis and intestinal microbiota disturbance to attenuate liver fibrosis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119571. [PMID: 40023344 DOI: 10.1016/j.jep.2025.119571] [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: 11/17/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanghuangporus, the dried fruiting body of Sanghuangporus vaninii (Ljub) L.W.Zhou et Y.C.Dai. As the main species of Sanghuang, it has been well-known and used commonly as a traditional medicinal and edible macrofungi for thousands of years in many countries, including China, Korea and Japan. Although it has good hepatoprotective activity, its potential efficacy and mechanism on liver fibrosis remain elusive. AIM OF THE STUDY Total Sanghuangporus vaninii extract (TSH) was prepared by ethanol extraction to investigate its chemical components and to conduct an initial assessment of its efficacy and underlying mechanism in a murine model of liver fibrosis. MATERIALS AND METHODS The chemical components of TSH were initially analyzed by UHPLC-Q-Orbitrap HRMS. To elucidate the effects of TSH, an in vivo model of fibrosis was established in mice using carbon tetrachloride (CCl4), followed by assessments of serum liver function and histopathological analysis. Besides, indicators related to liver fibrosis, hepatic stellate cells (HSCs) activation, inflammation response and ferroptosis related indicators were detected by western blotting, immunohistochemistry and real-time quantitative PCR (RT-qPCR) analysis. Additionally, the 16S rDNA sequencing and untargeted metabolomics analysis of intestinal microbiota were employed to investigating the role of TSH in gut microbiome. In vitro, the human hepatocyte line L02 was stimulated with erastin and treated with or without TSH to elucidate its underlying mechanism. RESULTS The administration of TSH significantly improved serum indicators of liver injury in CCl4-induced fibrosis mice, reduced HSCs activation and collagen deposition, while inhibiting the expressions of transforming growth factor-β1(TGF-β1)/Smad signaling pathway. Notably, TSH treatment attenuated hepatocyte ferroptosis and lipid peroxidation both in vivo and in vitro, as evidenced by a marked decrease in liver iron and malondialdehyde (MDA) contents. In particular, TSH was demonstrated to activate the nuclear factor erythroid 2-related factor 2 (Nrf2)-glutathione peroxidase 4 (GPX4) signaling pathway, thereby protecting hepatocytes from ferroptosis with a particular enhancement of Nrf2 nuclear transcription. Furthermore, TSH influenced gut microbiota composition and ameliorated intestinal metabolic disorders. The increased abundance of Parasutterella and Olsenellas due to TSH treatment was significantly positively correlated with elevated phosphatidylcholines involved in linoleic acid metabolism, and negatively correlated with the reduction of fatty acyls. And the enrichment of intestinal linoleic acid metabolism presented a negative correlation in liver fibrosis biomarkers. CONCLUSIONS Our findings indicate that the TSH treatment exerts a significantly protective effect on CCl4-induced mice by ameliorating hepatic injury and ferroptosis damage, inhibiting HSCs activation and collagen deposition, and remodeling gut microbiota homeostasis and metabolic imbalance. Notably, TSH attenuated hepatocyte ferroptosis in liver fibrosis and exhibited upregulation of the Nrf2-GPX4 signaling pathway. Furthermore, TSH could enrich the abundance of Parasutterella and Olsenellas, which may contribute to intestinal linoleic acid metabolism, thereby contributing to the reduction of liver fibrosis damage. Our study provides more effective and unreported evidence of TSH in anti-fibrosis activity.
Collapse
Affiliation(s)
- Siqi Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xingxing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qiuying Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Rongsheng Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lumeng Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Anna Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qun Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Zhun Xiao
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Shengsheng Li
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Jianjun Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
132
|
Hu Y, Ni X, Chen Q, Qu Y, Chen K, Zhu G, Zhang M, Xu N, Bai X, Wang J, Ma Y, Luo Q, Cai K. Predicting diabetic kidney disease with serum metabolomics and gut microbiota. Sci Rep 2025; 15:12179. [PMID: 40204798 PMCID: PMC11982385 DOI: 10.1038/s41598-025-91281-9] [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: 10/04/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
This study aims to identify biomarkers for reliably predicting diabetic kidney disease (DKD), systematically characterize serum metabolites and gut microbiota in DKD patients, and investigate the correlation between differential serum metabolites and gut microbiota. From September 2021 to January 2023, 90 subjects were recruited: 30 with DKD, 30 with type 2 diabetes mellitus (T2DM), and 30 normal controls (NCs). Serum metabolites, including 180 different metabolites, were analyzed using untargeted metabolomics UPLC-MS/MS, and gut microbiota were assessed via 16S rRNA sequencing. Differential metabolites were identified through univariate (t-test or Mann-Whitney U-test, P < 0.05) and multivariate analyses (OPLS-DA model, VIP > 1, P < 0.05), followed by selection using the least absolute shrinkage and selection operator (LASSO). The selected overlapping serum metabolites, along with DKD-associated differential gut microbiota, were used to develop a logistic regression prediction model for DKD based on six markers. In the DKD group compared to the DM and NC groups, 39 and 60 differential serum metabolites were identified, respectively (VIP > 1, P < 0.01). Among these, 36 serum metabolites, including alpha-Hydroxyisobutyric acid, were significantly elevated in DKD patients compared to those with DM. Of these, 28 metabolites showed a negative correlation with estimated glomerular filtration rate (eGFR), while 29 showed a positive correlation with urine albumin creatinine ratio (UACR). Patients with DKD were further categorized into subgroups (DKD middle and DKD early) based on eGFR (eGFR < 90 ml/min/1.73m2, eGFR ≥ 90 ml/min/1.73m2), revealing 23 differential metabolites. Dysbiosis of the gut microbiota was evident in DKD patients, with lower relative abundances of g_Prevotella and g_Faecalibacterium compared to the DM and NC groups. Subgroup analysis indicated decreased relative abundances of g_Prevotella and g_Faecalibacterium in the DKD middle group, along with a decrease in g_Klebsiella compared to the DKD early group, which correlated positively with DKD patients' eGFR. There were 11 common metabolites among the three groups of differential metabolites. Among these, three serum metabolites-imidazolepropionic acid, adipoylcarnitine, and 1-methylhistidine-were identified as predictive serum metabolic markers. Disease prediction models (logistic regression models) were constructed based on these three metabolites combined with three genera of bacteria. These models demonstrated strong discriminatory power for diagnosing patients with DKD compared to patients with DM (area under the receiver operating characteristic curve (AUROC) = 0.939 and precision-recall curve (AUPR) = 0.940). The models also effectively discriminated between patients with DKD and NCs (0.976, 0.973). This study revealed distinctive serum metabolites and gut microbiota in patients with DKD. It demonstrated the potential utility of three specific serum metabolites and three genera of bacteria in diagnosing patients with DKD and assessing their renal dysfunction.
Collapse
Affiliation(s)
- Yuyun Hu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xue Ni
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Qinghuo Chen
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yihui Qu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Kanan Chen
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Gaohui Zhu
- Department of Endocrinology, Ningbo Zhenhai Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang, China
| | - Minqiao Zhang
- Department of Nephrology, The First People's Hospital of Xiangshan, Ningbo, 315700, China
| | - Ningjie Xu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xu Bai
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Jing Wang
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yanhong Ma
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Luo
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Kedan Cai
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China.
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
133
|
Ba J, Zheng C, Lai Y, He X, Pan Y, Zhao Y, Xie H, Wu B, Deng X, Wang N. High matrix stiffness promotes senescence of type II alveolar epithelial cells by lysosomal degradation of lamin A/C in pulmonary fibrosis. Respir Res 2025; 26:128. [PMID: 40205454 PMCID: PMC11984030 DOI: 10.1186/s12931-025-03201-0] [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: 02/14/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Cellular senescence is one of the key steps in the progression of pulmonary fibrosis, and the senescence of type II alveolar epithelial cells (AEC IIs) may potentially accelerate the progression of pulmonary fibrosis. However, the molecular mechanisms underlying cellular senescence in pulmonary fibrosis remain unclear. METHODS The researchers first conducted in vitro experiments to investigate whether AEC IIs cultured on high matrix stiffness would lead to cellular senescence. Next, samples from mouse pulmonary fibrosis models and clinical idiopathic pulmonary fibrosis (IPF) patients were tested to observe extracellular matrix deposition, lamin A/C levels, and cellular senescence status in lung tissue. Construct lamin A/C knockdown and overexpression systems separately in AEC IIs, and observe whether changes in lamin A/C levels lead to cellular senescence. Further explore the degradation mechanism of lamin A/C using protein degradation inhibitors. RESULTS In vitro experiments have found that high matrix stiffness promotes senescence of AEC IIs. In a mouse model of pulmonary fibrosis, AEC IIs were found to exhibit significant cellular senescence on day 21. In clinical IPF samples, it was found that senescent cells expressed low levels of lamin A/C. In the lamin A/C SiRNA knockdown system, it was further confirmed that AEC IIs with low levels of lamin A/C are more prone to cellular senescence. Under high matrix stiffness, lamin A/C in AEC IIs is degraded through the autophagy lysosome pathway. The use of chloroquine can effectively alleviate cellular senescence. CONCLUSIONS High matrix stiffness degrades lamin A/C in pulmonary fibrosis through lysosomal degradation pathways, promoting AEC II senescence. Inhibition the degradation of lamin A/C could alleviate AEC II senescence.
Collapse
Affiliation(s)
- Junhui Ba
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Changyu Zheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yimei Lai
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Xin He
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yuxi Pan
- Department of Oncology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yanqiu Zhao
- Shenzhen Samii Medical Center, Shenzhen, Guangdong Province, China
| | - Huihui Xie
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Benquan Wu
- Department of Medical Intensive Care Unit, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - Xiao Deng
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China.
| | - Nan Wang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong Province, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China.
| |
Collapse
|
134
|
Volarevic V, Randall Harrell C, Arsenijevic A, Djonov V. An Interplay Between Pericytes, Mesenchymal Stem Cells, and Immune Cells in the Process of Tissue Regeneration. Anal Cell Pathol (Amst) 2025; 2025:4845416. [PMID: 40241723 PMCID: PMC12003036 DOI: 10.1155/ancp/4845416] [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: 09/16/2023] [Revised: 02/10/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Immediately after injury, damaged cells elicit tissue regeneration, a healing process that enables optimal renewal and regrowth of injured tissues. Results obtained in a large number of experimental studies suggested that the cross talk between pericytes, mesenchymal stem cells (MSC), tissue-resident stem cells, and immune cells has a crucially important role in the regeneration of injured tissues. Pericytes, MSCs, and immune cells secrete bioactive factors that influence each other's behavior and function. Immune cells produce inflammatory cytokines and chemokines that influence pericytes' migration, proliferation, and transition to MSC. MSC releases immunoregulatory factors that induce the generation of immunosuppressive phenotype in inflammatory immune cells, alleviating detrimental immune responses in injured tissues. MSC also produces various growth factors that influence the differentiation of tissue-resident stem cells into specific cell lineages, enabling the successful regeneration of injured tissues. A better understanding of molecular mechanisms that regulate crosstalk between pericytes, MSC, and immune cells in injured tissues would enable the design of new therapeutic approaches in regenerative medicine. Accordingly, in this review paper, we summarized current knowledge related to the signaling pathways that are involved in the pericytes' activation, pericytes-to-MSC transition, differentiation of tissue-resident stem cells, and MSC-dependent modulation of immune cell-driven inflammation, which are crucially responsible for regeneration of injured tissues.
Collapse
Affiliation(s)
- Vladislav Volarevic
- Center for Harm Reduction of Biological and Chemical Hazards, Department of Genetics and Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, Kragujevac, Serbia
| | - Carl Randall Harrell
- Regenerative Processing Plant, LLC 34176, US Highway 19 N, Palm Harbor, Florida, USA
| | - Aleksandar Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Department of Genetics and Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, Kragujevac, Serbia
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2 3012, Bern, Switzerland
| |
Collapse
|
135
|
Su L, Sun Q, Li Y, Alvarez JF, Tao B, Zhang G, Gu Y, Hanudel MR, Espinoza A, Zhang L, Pan C, Hilser JR, Hartiala JA, Li S, Pellegrini M, Allayee H, Lusis AJ, Deb A. Collagen V regulates renal function after kidney injury and can be pharmacologically targeted to enhance kidney repair in mice. Sci Transl Med 2025; 17:eads7714. [PMID: 40203084 DOI: 10.1126/scitranslmed.ads7714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/03/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Kidney fibrosis determines clinical outcomes in individuals with chronic kidney disease (CKD). The stoichiometric ratio of collagens in renal scar differs from that of healthy kidney extracellular matrix (ECM), but the functional importance of altered collagen types in injured kidneys remains unclear. Using human population studies, we show that circulating protein and renal mRNA amounts of collagen V A1 (COL5A1) exhibited associations with kidney disease and incident CKD risk. We show that Col5a1 regulates the degree of postinjury fibrosis and renal function. Mice with conditionally knocked out Col5a1 (Col5a1 CKO) exhibited decreased renal function and greater renal fibrosis after dietary adenine- or ureteric obstruction-mediated kidney injury. Renal fibroblasts in Col5a1 CKO animals up-regulated the profibrotic αvβ3 integrin. Inhibition of αvβ3 signaling with a small molecule, cilengitide, rescued postinjury renal function in Col5a1 CKO animals. Using the hybrid mouse diversity panel that comprises 100 diverse inbred strains of mice, we observed that gene expression of Col5a1 after injury exhibited genetic variation across 100 strains. Strains with low Col5a1 expression after injury exhibited worse renal function compared with animals that had higher degrees of expression. We next measured Col5a1 expression in peripheral blood mononuclear cells in mice to identify nonresponder strains that did not have increased Col5a1 expression after kidney injury. We observed that administration of cilengitide in nonresponder strains significantly rescued postinjury renal fibrosis and function. These studies point to the feasibility of precision medicine approaches to target Col5a1 for enhancing renal repair.
Collapse
Affiliation(s)
- Lianjiu Su
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qihao Sun
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yusheng Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Juan Felipe Alvarez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yiqian Gu
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark R Hanudel
- Department of Pediatric Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Espinoza
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linlin Zhang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - James R Hilser
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jaana A Hartiala
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hooman Allayee
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
136
|
Wu S, Bao H, Bau T. Integrated metabolomics and transcriptomics to reveal the anti-tumor mechanisms of Sanghuangporus mongolicus ethyl acetate extract in H22 tumor-bearing mice. Fitoterapia 2025; 183:106536. [PMID: 40210199 DOI: 10.1016/j.fitote.2025.106536] [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: 02/05/2025] [Revised: 03/24/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
This study aimed to systematically evaluate the anti-tumor efficacy of petroleum ether, ethyl acetate (EAE), and water extracts from Sanghuangporus mongolicus and decipher the molecular mechanisms of the most efficacious extract - EAE. Using H22 tumor-bearing mice, high-dose EAE (37.72 mg/kg) exhibited the highest tumor inhibition rate (75.14 %) without toxicity. Histopathology examination demonstrated that EAE effectively mitigated tumor progression and multi-organ damage, whereas ELISA and serum biochemical assays indicated modulated levels of immune mediators (IL-6, IL-2, IFN-γ, TNF-α, and VEGF) and restored serum levels of hepatic (ALT, AST) and renal (BUN, Cr, UA) functional markers. Integrated transcriptomics and metabolomics demonstrated that EAE suppressed tumor growth via multi-target regulation involving immune responses, biosynthesis of amino acids, and mitochondrial apoptosis pathways. Western blotting validated EAE upregulated pro-apoptotic cleaved caspase-3, caspase-3, Bax, and TNF-α (p < 0.01 vs. model group (MG)), upregulated immune-related protein JCHAIN, MZB1 (p < 0.01 vs. MG), downregulated anti-apoptotic Bcl-2 (p < 0.01 vs. MG). LC-MS identified 33 EAE compounds, with 32 showing strong binding (ΔG ≤ -5.0 kcal/mol) to core targets, JCHAIN and MZB1, via molecular docking. Phellibaumin C, inoscavin A, and phelligridin D exhibited the highest binding affinities. This study provides a multi-target mechanistic framework for developing S. mongolicus EAE as a natural anti-hepatocellular carcinoma (HCC) agent.
Collapse
Affiliation(s)
- Sitegele Wu
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun, Jilin Province 130118, China; College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
| | - Haiying Bao
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun, Jilin Province 130118, China; College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province 130118, China
| | - Tolgor Bau
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun, Jilin Province 130118, China; College of Mycology, Jilin Agricultural University, Changchun, Jilin Province 130118, China.
| |
Collapse
|
137
|
Chen S, Huang Y, Qu Z. Telitacicept monotherapy for refractory idiopathic membranous nephropathy: a case report and literature review. Front Med (Lausanne) 2025; 12:1571616. [PMID: 40265191 PMCID: PMC12011803 DOI: 10.3389/fmed.2025.1571616] [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: 02/05/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Background Patients with refractory membranous nephropathy (MN) face risks of progressive renal decline and end-stage renal disease (ESRD), with limited treatment efficacy. Telitacicept, a novel humanized recombinant fusion protein effective in lupus nephritis and immunoglobulin A nephropathy (IgAN), has few reports on its use in refractory MN. Case presentation In May 2023, an 82-year-old man was admitted to Shaoxing Second Hospital with bilateral lower extremity edema. A renal biopsy confirmed idiopathic membranous nephropathy (IMN). Standard therapies, including glucocorticoids (GC), cyclophosphamide (CYC), tacrolimus (TAC), and rituximab (RTX), were ineffective. He developed steroid-induced diabetes and acute renal failure during treatment. Complete proteinuria remission was achieved with telitacicept monotherapy. The patient is under ongoing clinical follow-up. Conclusion Telitacicept holds promise as a potential second-line therapy for refractory MN when conventional treatments prove ineffective. However, due to the current lack of robust evidence supporting its use in IMN, further research is warranted to establish its clinical efficacy and safety.
Collapse
Affiliation(s)
- Shucheng Chen
- Department of Endocrinology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Yiqi Huang
- Department of Nephrology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Zhongjie Qu
- Department of Nephrology, The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, Zhejiang, China
| |
Collapse
|
138
|
Zhang C, Bai K, Li D. Downregulation of S100 calcium-binding A4 (S100A4) ameliorates hepatic fibrosis via regulating Wnt/β-catenin signaling pathway. Eur J Histochem 2025; 69:4186. [PMID: 40223805 PMCID: PMC12051413 DOI: 10.4081/ejh.2025.4186] [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: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
S100 calcium-binding protein A4 (S100A4), a fibrosis-associated calcium-binding protein, has been implicated in fibrotic progression across multiple organs. Activation of the Wnt/β-catenin signaling pathway is a critical driver of hepatic fibrosis, yet the mechanistic role of S100A4 in this context remains poorly defined. This study investigated the regulatory role of S100A4 in hepatic fibrosis in vitro and in vivo. Hepatic stellate cells (HSCs) were treated with TGF-β to induce fibrotic activation, and S100A4 expression was silenced using shRNA. A carbon tetrachloride (CCl₄)-induced murine hepatic fibrosis model was employed for in vivo validation. Fibrotic markers, including collagen I, fibronectin, and α-smooth muscle actin (α-SMA), were assessed via qRT-PCR, Western blotting, immunofluorescence, and immunohistochemistry. Liver histopathology and function were evaluated using Masson trichrome staining, hematoxylin-eosin staining, and serum ALT/AST assays. In vitro experiments demonstrated that TGF-β treatment upregulated S100A4 expression in HSCs, while S100A4 silencing suppressed HSC activation, extracellular matrix (ECM) deposition, and Wnt/β-catenin signaling. In vivo, S100A4 downregulation attenuated CCl₄-induced hepatic fibrosis, reduced collagen accumulation, improved liver histology, and normalized serum ALT/AST levels. These findings indicate that S100A4 promotes hepatic fibrosis by activating the Wnt/β-catenin pathway, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
| | | | - Dexu Li
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
139
|
Dilger OB, Carstens MF, Bothun CE, Payne AN, Berry DJ, Sanchez-Sotelo J, Morrey ME, Thaler R, Dudakovic A, Abdel MP. Induction of cellular autophagy impairs TGF-β1-mediated extracellular matrix deposition in primary human knee fibroblasts. Bone Joint Res 2025; 14:331-340. [PMID: 40192622 PMCID: PMC11975063 DOI: 10.1302/2046-3758.144.bjr-2024-0312.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Aims To evaluate the role of autophagy in primary knee fibroblasts undergoing myofibroblast differentiation as an in vitro model of arthrofibrosis, a complication after total knee arthroplasty characterized by aberrant intra-articular scar tissue formation and limited range of motion. Methods We conducted a therapeutic screen of autophagic-modulating therapies in primary human knee fibroblasts undergoing transforming growth factor-beta 1 (TGF-β1)-mediated myofibroblast differentiation. Autophagy was induced pharmacologically with rapamycin or by amino acid deprivation. Picrosirius red staining was performed to quantify collagen deposition. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were conducted to evaluate fibrotic gene expression levels. Results Rapamycin, an mTOR complex 1 (mTORC1) inhibitor and autophagy inducer, reduced TGF-β1-mediated collagen deposition. Interestingly, we simultaneously report that myofibrogenic genes, including ACTA2, were highly upregulated following rapamycin-TGF-β1 treatment. When autophagy was induced through amino acid deprivation, we demonstrated suppressed extracellular matrix levels, fibrotic gene expression (e.g. ACTA2), and SMAD2 phosphorylation levels in TGF-β1-stimulated fibroblasts. Conclusion Our findings demonstrate that the induction of cellular autophagy suppresses TGF-β1-induced collagen deposition in primary human knee fibroblasts. Taken together, these data suggest that cellular autophagy may be prophylactic against the pathogenesis of arthrofibrosis.
Collapse
Affiliation(s)
- Oliver B. Dilger
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mason F. Carstens
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Cole E. Bothun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ashley N. Payne
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J. Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mark E. Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P. Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
140
|
Liu Y, Zhou Y, Zhang H, Zhao K, Yang D. Gut-lung Axis mediates asthma pathogenesis: Roles of dietary patterns and their impact on the gut microbiota. Exp Mol Pathol 2025; 142:104964. [PMID: 40194490 DOI: 10.1016/j.yexmp.2025.104964] [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: 10/26/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
The gut-lung axis, a vital signaling network linking the gastrointestinal and pulmonary systems, regulates immune responses and the progression of respiratory diseases. Nutritional components can modulate the gut microbiome and regulate the synthesis of critical intestinal microbial metabolites, which are essential for maintaining immune homeostasis and supporting respiratory health. Conversely, poor dietary habits exacerbate asthma and other respiratory conditions through the modulation of systemic inflammation and immune responses. Dietary interventions, such as the Mediterranean diet, are reported to restore microbial balance and improve respiratory health by increasing the production of anti-inflammatory metabolites, potentiating immune responses, and preserving epithelial barrier integrity. In contrast, Western dietary patterns, which are characterized by high fat and low fiber intake, disrupt microbial diversity, resulting in increased levels of pro-inflammatory metabolites that aggravate airway inflammation and asthma severity. This review aimed to elucidate the mechanisms underlying the regulatory effects of gut microbes and their metabolites on asthma. Additionally, previous findings related to the gut-lung axis have been summarized, providing insights into potential therapeutic strategies for asthma management.
Collapse
Affiliation(s)
- Yanbo Liu
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Zhou
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haoyue Zhang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaixuan Zhao
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Yang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
141
|
Gencler A, Celik H, Demir A. Evaluating serum S-Equol, indoxyl sulfate, and TMAO in predicting urinary stones in children: a prospective study. Urolithiasis 2025; 53:68. [PMID: 40186630 PMCID: PMC11972205 DOI: 10.1007/s00240-025-01737-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
Gut microbiota is vital in maintaining health and has been implicated in urinary stone disease. Patients with and without stones have different microbial compositions. In this context, we assessed serum levels of S-equol, indoxyl sulfate (IS), and trimethylamine N-oxide (TMAO), which are metabolites thought to be associated with gut microbiota, and their prognostic values in predicting stone formation in children with urinary stone disease. The study population consisted of children aged between one month and 18 years with urinary stone disease. The patient group consisted of 44 children with urinary stone disease, and the control group consisted of 44 healthy children who were matched with the patient group in terms of age and gender. The study's primary outcomes were the differences between the groups in serum metabolite levels. Serum S-equol and TMAO levels were significantly lower in the patient group than in the control group. There was no significant difference between the groups in serum IS levels. There were also no significant correlations between serum metabolite levels and age in either group. Children with urinary stone disease had significantly lower serum S-equol and TMAO levels than healthy control subjects, suggesting a possible link between these metabolites and stone formation.
Collapse
Affiliation(s)
- Aylin Gencler
- Department of Pediatric Nephrology, Harran University Faculty of Medicine, Şanlıurfa, Turkey.
| | - Hakim Celik
- Department of Medical Physiology, Harran University Faculty of Medicine, Şanlıurfa, Turkey
| | - Abit Demir
- Department of Pediatrics, Harran University Faculty of Medicine, Şanlıurfa, Turkey
| |
Collapse
|
142
|
Jagua-Gualdrón A, García-Reyes NA, Africano-Lopez HL. Apitherapy for drug-induced kidney disease: a narrative review on its mechanisms. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025:jcim-2025-0082. [PMID: 40178599 DOI: 10.1515/jcim-2025-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVES The use of medications for the treatment of various diseases often results in kidney damage. Apitherapy is a natural therapeutic tool with potential utility for this purpose. This narrative review analyzes and summarizes the scientific evidence on the use of apitherapy in drug-induced kidney disease. CONTENT This review summarizes and analyzes recent advances in drug-induced kidney disease and explores, based on the available scientific evidence, how apitherapy can modify these mechanisms and be utilized for prevention and treatment. SUMMARY Apitherapy (the complementary and integrative use of beehive products) is a potentially useful therapeutic system for the treatment of various diseases. This review examines the preclinical and clinical evidence available regarding its potential use in drug-induced kidney disease. OUTLOOK Apitherapy has effects on various pathophysiological mechanisms of drug-induced kidney disease, including oxidative stress, inflammation, decreased renal blood flow, glomerular damage, increased membrane permeability, activity of the renin-angiotensin-aldosterone axis, mitochondrial dysfunction, and apoptosis. Further studies in humans are needed to evaluate its efficacy in the clinical setting, but the available evidence is promising.
Collapse
Affiliation(s)
- Andrés Jagua-Gualdrón
- National University of Colombia, Bogotá D.C., Colombia
- International Institute for Complementary and Alternative Medicine-IIMAN, Bogotá D.C., Colombia
- International College of Apitherapy, Bogotá D.C., Colombia
| | - Nicolai Andrés García-Reyes
- National University of Colombia, Bogotá D.C., Colombia
- International Institute for Complementary and Alternative Medicine-IIMAN, Bogotá D.C., Colombia
| | - Holman Leonardo Africano-Lopez
- Fundación Universitaria de Ciencias de la Salud, Sociedad de Cirugía de Bogotá, Hospital San José Sede Centro, Bogotá D.C., Colombia
| |
Collapse
|
143
|
Sun M, Zhang W, Tian C, Wang R, Liu W, Li Y, Lv Y, Wang Z. A Novel Classification Model Based on Hyperspectral Imaging for Predicting Response to Tacrolimus in Patients With Primary Membranous Nephropathy. JOURNAL OF BIOPHOTONICS 2025:e70025. [PMID: 40181509 DOI: 10.1002/jbio.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 04/05/2025]
Abstract
At present, the research to predict the efficacy of tacrolimus (TAC) mainly focuses on serological indexes and urine analysis. Because these indicators are affected by many factors, they cannot accurately predict the therapeutic effect of primary membranous nephropathy (PMN) patients. In this study, a novel classification model (RCN) based on hyperspectral imaging combined with one-dimensional convolutional neural networks (1D CNN) and relevance vector machine (RVM) was proposed for predicting patients' response to TAC. Based on the treatment outcomes of corticosteroids combined with TAC, the patients were divided into a remission group and a nonremission group. Through the analysis of hyperspectral data of pathological slices of patients in both the remission group and the nonremission group, the research results show that the model can effectively extract key features from the spectral data and achieve high classification performance, and it can predict the therapeutic effect of TAC in PMN patients.
Collapse
Affiliation(s)
- Meijuan Sun
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
| | - Wenqiang Zhang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
| | - Chongxuan Tian
- School of Control Science and Engineering, Shandong University, Jinan, China
| | - Ruiyang Wang
- School of Radiology, Shandong First Medical University, Taian, China
| | - Wen Liu
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
| | - Yang Li
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
| | - Yang Lv
- Marine and Freshwater Biology, College of Life Sciences, University of Glasgow, Glasgow, Scotland
| | - Zunsong Wang
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Jinan, China
| |
Collapse
|
144
|
Schneider S, Biggerstaff D, Barber TM. Dietary Guidelines Post Kidney Transplant: Is This the Missing Link in Recovery and Graft Survival? Transpl Int 2025; 38:14288. [PMID: 40248508 PMCID: PMC12004285 DOI: 10.3389/ti.2025.14288] [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: 01/02/2025] [Accepted: 03/11/2025] [Indexed: 04/19/2025]
Abstract
The physiology of a transplanted kidney is affected from the moment it is separated from the donor. The risk of complications arising from surgery are highly associated with ischemic-reperfusion injury (IRI) due to the effects of hypoxia and oxidative stress during the procurement, preservation and reperfusion procedures. Hypoxia promotes the formation of reactive oxygen species (ROS) and it seems apparent that finding ways of optimising the metabolic milieu for the transplanted kidney would improve recovery and graft survival. Studies have demonstrated the benefits of nutrition and antioxidant compounds in mitigating the disturbance of energy supply to cells post-transplant and at improving long-term graft survival. Particularly in patients who may be nutritionally deficient following long-term dialysis. Despite the high incidence of allograft failure, a search of the literature and grey literature reveals no medical nutriti on therapy guidelines on beneficial nutrient intake to aid transplant recovery and survival. This narrative review aims to summarise current knowledge of specific macro and micronutrients and their effect on allograft recovery and survival in the perioperative period, up to 1-year post transplant, to optimise the metabolic environment and mitigate risk to graft injury.
Collapse
Affiliation(s)
- Suzanne Schneider
- Directorate Applied Health, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Deborah Biggerstaff
- Directorate Applied Health, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Thomas M. Barber
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| |
Collapse
|
145
|
Cheng S, Wang H. Aging and atrial fibrillation: Role of telomere dysfunction. Chin Med J (Engl) 2025:00029330-990000000-01507. [PMID: 40176558 DOI: 10.1097/cm9.0000000000003550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Indexed: 04/04/2025] Open
Affiliation(s)
- Sijun Cheng
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, China Medical University, Shenyang, Liaoning 110000, China
| | | |
Collapse
|
146
|
Gill SK, Gomer RH. Translational Regulators in Pulmonary Fibrosis: MicroRNAs, Long Non-Coding RNAs, and Transcript Modifications. Cells 2025; 14:536. [PMID: 40214489 PMCID: PMC11988943 DOI: 10.3390/cells14070536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Fibrosing disorders including idiopathic pulmonary fibrosis (IPF) are progressive irreversible diseases, often with poor prognoses, characterized by the accumulation of excessive scar tissue and extracellular matrix. Translational regulation has emerged as a critical aspect of gene expression control, and the dysregulation of key effectors is associated with disease pathogenesis. This review examines the current literature on translational regulators in IPF, focusing on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and RNA transcript modifications including alternative polyadenylation and chemical modification. Some of these translational regulators potentiate fibrosis, and some of the regulators inhibit fibrosis. In IPF, some of the profibrotic regulators are upregulated, and some of the antifibrotic regulators are downregulated. Correcting these defects in IPF-associated translational regulators could be an intriguing avenue for therapeutics.
Collapse
Affiliation(s)
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
147
|
Ratan Y, Rajput A, Pareek A, Pareek A, Singh G. Comprehending the Role of Metabolic and Hemodynamic Factors Alongside Different Signaling Pathways in the Pathogenesis of Diabetic Nephropathy. Int J Mol Sci 2025; 26:3330. [PMID: 40244213 PMCID: PMC11989741 DOI: 10.3390/ijms26073330] [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: 02/07/2025] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
Diabetic nephropathy (DN) is a progressive microvascular disorder of diabetes that contributes as a primary reason for end-stage renal disease worldwide. The pathological hallmarks of DN include diffuse mesangial expansion, thicker basement membrane of glomeruli, and arteriole hyalinosis. Hypertension and chronic hyperglycemia are the primary risk factors contributing to the occurrence of DN. The complex pathophysiology of DN involves the interplay amongst metabolic and hemodynamic pathways, growth factors and cytokines production, oxidative stress, and ultimately impaired kidney function. Hyperglycemia-induced vascular dysfunction is the main pathological mechanism that initiates DN. However, several other pathogenic mechanisms, such as oxidative stress, inflammatory cell infiltration, and fibrosis, contribute to disease progression. Different vasoactive hormone processes, including endothelin and renin-angiotensin, are activated as a part of the pathophysiology of DN, which also involves increased intraglomerular and systemic pressure. The pathophysiology of DN will continue to be better understood because of recent developments in genomics and molecular biology, but attempts to develop a comprehensive theory that explains all existing cellular and biochemical pathways have been thwarted by the disease's multifactorial nature. This review extensively discusses the current understanding regarding the metabolic and hemodynamic pathological mechanisms, along with other signaling pathways and molecules responsible for the pathogenesis of DN. This work will encourage a greater in-depth understanding and investigation of the present status of the biochemical mechanistic processes underlying the pathogenesis of DN, which may assist in the determination of different biomarkers and help in the design and development of novel drug candidates in the near future.
Collapse
Affiliation(s)
- Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (Y.R.); (A.R.); (A.P.); (A.P.)
| | - Aishwarya Rajput
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (Y.R.); (A.R.); (A.P.); (A.P.)
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (Y.R.); (A.R.); (A.P.); (A.P.)
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (Y.R.); (A.R.); (A.P.); (A.P.)
| | | |
Collapse
|
148
|
Gao X, Fan X, Yu X, Wang R, Zhang B, Li Y, Liu X, Yang Y. p66shc exacerbates the progression of obstructive nephropathy through apoptosis, mitochondrial damage, and EMT. J Pediatr Urol 2025:S1477-5131(25)00158-5. [PMID: 40234179 DOI: 10.1016/j.jpurol.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Many factors contribute to hydronephrosis, ultimately resulting in renal fibrosis and even deterioration of renal function. This study investigated the pathogenic role of p66shc, a redox-regulatory protein, in hydronephrosis-induced renal injury. OBJECTIVE This study focused on the mechanism of p66shc in renal fibrosis associated with obstructive nephropathy. METHODS The expression of p66shc was found in kidney samples from pediatric hydronephrosis patients. A complete unilateral ureteral obstruction (CUUO) model was established in neonatal mice to recapitulate hydronephrotic progression. Cell proliferation, apoptosis, reactive oxygen species (ROS), mitochondrial damage, and degree of epithelial-mesenchymal transition (EMT) in renal tubular epithelial cells were studied following p66shc silencing and overexpression. We also investigated the therapeutic effects of silencing p66shc in vivo and carried out RNA sequencing after overexpressing p66shc in cells. RESULTS p66shc inhibited renal tubular epithelial cell growth, exacerbated cell oxidative and mitochondrial damage, and promoted cell apoptosis and EMT. Silencing its expression in vivo could efficiently reduce renal fibrosis. Combined with RNA sequencing, we analyzed the potential molecular mechanisms of p66shc downstream. CONCLUSION p66shc enhances cell damage and the EMT process in obstructive nephropathy. Suppressing the expression of p66shc is one potential strategy for mitigating renal fibrotic progression.
Collapse
Affiliation(s)
- Xilin Gao
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xu Fan
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaohan Yu
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Rui Wang
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Buzhou Zhang
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yanqiu Li
- Department of Nephrology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Xin Liu
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China.
| | - Yi Yang
- Department of Pediatric Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
149
|
Lai Y, Zhu Y, Zhang X, Ding S, Wang F, Hao J, Wang Z, Shi C, Xu Y, Zheng L, Huang W. Gut microbiota-derived metabolites: Potential targets for cardiorenal syndrome. Pharmacol Res 2025; 214:107672. [PMID: 40010448 DOI: 10.1016/j.phrs.2025.107672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
The characteristic of cardiorenal syndrome (CRS) is simultaneous damage to both the heart and kidneys. CRS has caused a heavy burden of mortality and incidence rates worldwide. The regulation of host microbiota metabolism that triggers heart and kidney damage is an emerging research field that promotes a new perspective on cardiovascular risk. We summarize current studies from bench to bedside of gut microbiota-derived metabolites to better understand CRS in the context of gut microbiota-derived metabolites. We focused on the involvement of gut microbiota-derived metabolites in the pathophysiology of CRS, including lipid and cholesterol metabolism disorders, coagulation abnormalities and platelet aggregation, oxidative stress, endothelial dysfunction, inflammation, mitochondrial damage and energy metabolism disorders, vascular calcification and renal fibrosis, as well as emerging therapeutic approaches targeting CRS metabolism in gut microbiota-derived metabolites which provides an innovative treatment approach for CRS to improve patient prognosis and overall quality of life.
Collapse
Affiliation(s)
- Yuchen Lai
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yujie Zhu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Xihui Zhang
- Department of Blood Purification, General Hospital of Central Theater Command(Hankou Campus), No.68, Huangpu Avenue, Wuhan, 430010, China
| | - Shifang Ding
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China
| | - Fang Wang
- Department of Blood Purification, General Hospital of Central Theater Command(Hankou Campus), No.68, Huangpu Avenue, Wuhan, 430010, China
| | - Jincen Hao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Zhaomeng Wang
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China
| | - Congqi Shi
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yongjin Xu
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China.
| | - Wei Huang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China.
| |
Collapse
|
150
|
Zhang H, Hong Z, Jiang Z, Hu W, Hu J, Zhu R. miR-29b-3p Affects the Hypertrophy of Ligamentum Flavum in Lumbar Spinal Stenosis and its Mechanism. Biochem Genet 2025; 63:1824-1838. [PMID: 38625592 DOI: 10.1007/s10528-024-10811-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
To explore the effect of miR-29b-3p on fibrosis and hypertrophy of ligamentum flavum (LF) in lumbar spinal stenosis (LSS) and its underlying mechanism. Patients with LSS and lumbar disc herniation (LDH) (control) undergoing posterior lumbar laminectomy were included in this study. Human LF samples were obtained for LF cell isolation, RNA, and protein extraction. Histomorphological analysis of LF was performed using hematoxylin-eosin (HE) staining. After isolation, culture, and transfection of primary LF cells, different transfection groups were constructed: NC-mimic, miR-29b-3p-mimic, NC-inhibitor, and miR-29b-3p-inhibitor. Quantitative real time polymerase chain reaction (qRT-PCR) was performed to detect the expression of miR-29b-3p in LF and LF cells. Western blot analysis detected the protein expressions of P16 and CyclinD1. ELISA detected the protein expressions of TGF-β1, Smad2, Smad3, TLR4, Type I collagen, and Type III collagen. Finally, LF cell viability was detected using the Cell Counting Kit-8 (CCK8) assay. The thickness of LF was significantly thicker in the LSS group compared to the LDH group (p < 0.05), accompanied by a higher calcification degree, more fibroblasts, and a larger area of collagen fiber proliferation. miR-29b-3p expression was significantly lower in LSS-derived LF tissues and cells than in LDH-derived tissues and cells (both p < 0.05). Compared to the NC-mimic group, the miR-29b-3p-mimic group exhibited significantly higher miR-29b-3p expression, decreased protein expressions of Type I collagen, Type III collagen, TGF-β1, Smad2, Smad3, TLR4, P16, and CyclinD1, and inhibited LF cell proliferation (all p < 0.05). As expected, the miR-29b-3p-inhibitor group displayed contrasting expression patterns (all p < 0.05). Compared to the phosphate buffer saline (PBS) group, the Trimethylamine-N-Oxide (TMAO) group showed significantly increased expressions of TGF-β1, Smad2, Smad3, TLR4, Type I collagen, Type III collagen, P16, and CyclinD1, as well as enhanced LF cell proliferation (all p < 0.05). However, there was no significant difference between the TMAO group and the Ang II group (all p > 0.05). Upregulation of miR-29b-3p expression may play a role in improving LF fibrosis and hypertrophy in LSS by inhibiting P16 expression and suppressing the activation of the TGF-β/Smad signaling pathway. This finding offers new insights into future gene modification therapy for this patient population.
Collapse
Affiliation(s)
- Hongjie Zhang
- Department of Orthopedics, Dehong People's Hospital, Kunming Medical University Affiliated Dehong Hospital, Dehong, No.13 Yonghan Road, Mangshi District, 678400, China
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, No.10, Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Zhixiong Hong
- Department of Orthopedics, Dehong People's Hospital, Kunming Medical University Affiliated Dehong Hospital, Dehong, No.13 Yonghan Road, Mangshi District, 678400, China
| | - Zehua Jiang
- Department of Spine Surgery, Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300122, China
| | - Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300122, China
| | - Jiashao Hu
- Department of Orthopedics, Dehong People's Hospital, Kunming Medical University Affiliated Dehong Hospital, Dehong, No.13 Yonghan Road, Mangshi District, 678400, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300122, China.
| |
Collapse
|