51
|
Zhao BR, Hu XR, Wang WD, Zhou Y. Cardiorenal syndrome: clinical diagnosis, molecular mechanisms and therapeutic strategies. Acta Pharmacol Sin 2025; 46:1539-1555. [PMID: 39910210 PMCID: PMC12098865 DOI: 10.1038/s41401-025-01476-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025]
Abstract
As the heart and kidneys are closely connected by the circulatory system, primary dysfunction of either organ usually leads to secondary dysfunction or damage to the other organ. These interactions play a major role in the pathogenesis of a clinical entity named cardiorenal syndrome (CRS). The pathophysiology of CRS is complicated and involves multiple body systems. In early studies, CRS was classified into five subtypes according to the organs associated with the vicious cycle and the acuteness and chronicity of CRS. Increasing evidence shows that CRS is associated with a variety of pathological mechanisms, such as haemodynamics, neurohormonal changes, hypervolemia, hypertension, hyperuraemia and hyperuricaemia. In this review, we summarize the classification and currently available diagnostic biomarkers of CRS. We highlight the recently revealed molecular pathogenesis of CRS, such as oxidative stress and inflammation, hyperactive renin‒angiotensin‒aldosterone system, maladaptive Wnt/β-catenin signalling pathway and profibrotic TGF‒β1/Smad signalling pathway, as well as other pathogeneses, such as dysbiosis of the gut microbiota and dysregulation of noncoding RNAs. Targeting these CRS-associated signalling pathways has new therapeutic potential for treating CRS. In addition, various chemical drugs, natural products, complementary therapies, blockers, and agonists that protect against CRS are summarized. Since the molecular mechanisms of CRS remain to be elucidated, no single intervention has been shown to be effective in treating CRS. Pharmacologic therapies designed to block CRS are urgently needed. This review presents a critical therapeutic avenue for targeting CRS and concurrently illuminates challenges and opportunities for discovering novel treatment strategies for CRS.
Collapse
Affiliation(s)
- Bo-Rui Zhao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin-Rong Hu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Wei-Dong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| |
Collapse
|
52
|
Uranbileg B, Hoshino Y, Ezaka M, Kurano M, Uchida K, Yatomi Y, Ito N. Metabolism of sphingolipids in a rat spinal cord stenosis model. Biochem Biophys Rep 2025; 42:102025. [PMID: 40342530 PMCID: PMC12059668 DOI: 10.1016/j.bbrep.2025.102025] [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: 11/25/2024] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
Background Lumbar spinal canal stenosis (LSCS) plays a crucial role in neurogenic claudication and neuropathic pain. Recent studies suggest that changes in sphingolipid metabolism are linked to neuropathic pain. To explore the association between sphingolipids and LSCS, we measured the levels of sphingolipids and sphingolipid-associated molecules in an animal model of cauda equina compression (CEC), a typical type of LSCS. Methods By placing silicon blocks within the lumbar epidural space, CEC model were constructed in which motor disfunction had already been confirmed in our previous study. Quantitative measurements of various sphingolipids were conducted using LC-MS/MS in spinal cord and cerebrospinal fluid (CSF) samples on days 1, 7, and 28 following insertion of silicon blocks. Additionally, gene expression was analyzed in spinal cord tissue. Results In the CEC model, there was a significant increase ceramide levels in the CSF with upregulation of ceramide synthase 1 in the spinal cord tissue samples on day 1. Further, S1P levels in the CSF increased on day 7 and in the spinal cord significantly increased on day 28, and there was an increase in mRNA expression levels of sphingosine kinases (SphK)1 on days 1,7, and 28, while SphK2 on days 7 and 28. Regarding S1P receptors, there was an increase in mRNA expression levels of S1P1 on days 1,7, and 28 and S1P3 on day1. Conclusion The production and activation of the sphingolipid signaling pathway could play a pivotal role in neuropathic pain related to LSCS.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoko Hoshino
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Mariko Ezaka
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuko Ito
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
53
|
Yang MH, Basappa B, Deveshegowda SN, Ravish A, Mohan A, Nagaraja O, Madegowda M, Rangappa KS, Deivasigamani A, Pandey V, Lobie PE, Hui KM, Sethi G, Ahn KS. A novel drug prejudice scaffold-imidazopyridine-conjugate can promote cell death in a colorectal cancer model by binding to β-catenin and suppressing the Wnt signaling pathway. J Adv Res 2025; 72:615-632. [PMID: 39067696 DOI: 10.1016/j.jare.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
INTRODUCTION Globally, colorectal cancer (CRC) is the third most common type of cancer, and its treatment frequently includes the utilization of drugs based on antibodies and small molecules. The development of CRC has been linked to various signaling pathways, with the Wnt/β-catenin pathway identified as a key target for intervention. OBJECTIVES We have explored the impact of imidazopyridine-tethered chalcone-C (CHL-C) in CRC models. METHODS To determine the influence of CHL-C on apoptosis and autophagy, Western blot analysis, annexin V assay, cell cycle analysis, acridine orange staining, and immunocytochemistry were performed. Next, the activation of the Wnt/β-catenin signaling pathway and the anti-cancer effects of CHL-C in vivo were examined in an orthotopic HCT-116 mouse model. RESULTS We describe the synthesis and biological assessment of the CHL series as inhibitors of the viability of HCT-116, SW480, HT-29, HCT-15, and SNU-C2A CRC cell lines. Further biological evaluations showed that CHL-C induced apoptosis and autophagy in down-regulated β-catenin, Wnt3a, FZD-1, Axin-1, and p-GSK-3β (Ser9), and up-regulated p-GSK3β (Tyr216) and β-TrCP. In-depth analysis using structure-based bioinformatics showed that CHL-C strongly binds to β-catenin, with a binding affinity comparable to that of ICG-001, a well-known β-catenin inhibitor. Additionally, our in vivo research showed that CHL-C markedly inhibited tumor growth and triggered the activation of both apoptosis and autophagy in tumor tissues. CONCLUSION CHL-C is capable of inducing apoptosis and autophagy by influencing the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Min Hee Yang
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Suresha N Deveshegowda
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Akshay Ravish
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Arunkumar Mohan
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Omantheswara Nagaraja
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Mahendra Madegowda
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Kanchugarakoppal S Rangappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 169610, Singapore
| | - Vijay Pandey
- Shenzhen Bay Laboratory, Shenzhen 518055, China; Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Peter E Lobie
- Shenzhen Bay Laboratory, Shenzhen 518055, China; Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, 169610, Singapore.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
54
|
Gong J, Lu H, Li Y, Xu Q, Ma Y, Lou A, Cui W, Song W, Qu P, Chen Z, Quan L, Liu X, Meng Y, Li X. ACE2 shedding exacerbates sepsis-induced gut leak via loss of microbial metabolite 5-methoxytryptophan. MICROBIOME 2025; 13:136. [PMID: 40442816 PMCID: PMC12123736 DOI: 10.1186/s40168-025-02128-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 04/29/2025] [Indexed: 06/02/2025]
Abstract
BACKGROUND Sepsis, a critical organ dysfunction resulting from an aberrant host response to infection, remains a leading cause of mortality in ICU patients. Recent evidence suggests that angiotensin-converting enzyme 2 (ACE2) contributes to intestinal barrier function, the mechanism of which is yet to be explored. Additionally, alterations in intestinal microbiota and microbial metabolites could affect gut homeostasis, thus playing a potential role in modulating sepsis progression. RESULTS ACE2 shedding weakens the integrity of the intestinal barrier in sepsis. Mice deficient in ACE2 exhibited increased intestinal permeability and higher mortality rates post-operation compared to their wild-type counterparts. Notably, ACE2 deficiency was associated with distinct alterations in gut microbiota composition and reductions in protective metabolites, such as 5-methoxytryptophan (5-MTP). Supplementing septic mice with 5-MTP ameliorated gut leak through enhanced epithelial cell proliferation and repair. The PI3K-AKT-WEE1 signaling pathway was identified as a key mediator of the beneficial effects of 5-MTP administration. CONCLUSION ACE2 plays a protective role in maintaining intestinal barrier function during sepsis, potentially through modulation of the gut microbiota and the production of key metabolite 5-MTP. Our study enriched the mechanisms by which ACE2 regulates gut homeostasis and shed light on further applications. Video Abstract.
Collapse
Affiliation(s)
- Jiacheng Gong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haoyang Lu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuhan Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qihan Xu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuanyuan Ma
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Anni Lou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wanfu Cui
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weihua Song
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Peng Qu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhuoer Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Linghao Quan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xi Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510515, China.
| | - Ying Meng
- Department of Respiratory Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
55
|
Kumaran R, Abdalla MMI, Caszo BA, Somanath SD. Saliva urea nitrogen for detection of kidney disease in adults: A meta-analysis of diagnostic test accuracy. PLoS One 2025; 20:e0324251. [PMID: 40440346 PMCID: PMC12121763 DOI: 10.1371/journal.pone.0324251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Kidney disease affects millions globally, especially in low and middle-income countries where access to diagnostic testing is limited. Saliva urea nitrogen (SUN) has been proposed as a simple, non-invasive alternative to traditional serum-based diagnostics. OBJECTIVE This study aimed to evaluate the diagnostic accuracy of SUN for detecting kidney disease in adults through a systematic review and meta-analysis. METHODS This review adhered to the PRISMA-DTA guidelines. A comprehensive search of five databases was conducted without language or date restrictions. Study quality was assessed using the QUADAS-2 tool. STATA version 17 was used for analysis. A random-effects model was used to estimate pooled sensitivity, specificity, and diagnostic odds ratios (DOR). Subgroup analysis was conducted based on the reference test used (serum creatinine or blood urea nitrogen). Heterogeneity was assessed using the I² statistic, and meta-regression explored sources of heterogeneity. RESULTS Seven studies (n = 1,933) met the inclusion criteria. In the serum creatinine (sCr) subgroup (2 studies), SUN showed pooled sensitivity of 0.44 (95% CI: 0.38-0.49), specificity 0.96 (95% CI: 0.95-0.98), DOR 18.89 (95% CI: 15.19-23.57), and AUC ~ 0.90. In the blood urea nitrogen (BUN) subgroup (5 studies), sensitivity was 0.83 (95% CI: 0.69-0.91), specificity 0.88 (95% CI: 0.78-0.94), DOR 37 (95% CI: 15-91), and AUC 0.93. Heterogeneity was moderate in the BUN subgroup (bivariate I² = 51%), with 42% of variability attributed to threshold effects. Meta-regression identified study country (p = 0.01), and reference test used (p = 0.02) as contributors to heterogeneity in sensitivity, while comorbidity (p = 0.001) significantly affected specificity. CONCLUSION SUN shows high diagnostic specificity and a good overall accuracy, particularly when compared to BUN, and may serve as a practical non-invasive screening tool in low- resource settings. While heterogeneity was present, SUN remains a promising diagnostic alternative and warrants further validation in diverse clinical populations.
Collapse
Affiliation(s)
- Reyna Kumaran
- Human Biology Department, School of Medicine, IMU University (Formerly known as International Medical University), Kuala Lumpur, Malaysia
| | - Mona Mohamed Ibrahim Abdalla
- Human Biology Department, School of Medicine, IMU University (Formerly known as International Medical University), Kuala Lumpur, Malaysia
| | - Brinnell Annette Caszo
- Human Biology Department, School of Medicine, IMU University (Formerly known as International Medical University), Kuala Lumpur, Malaysia
| | - Sushela Devi Somanath
- Pathology Department, School of Medicine, IMU University (Formerly known as International Medical University), Kuala Lumpur, Malaysia
| |
Collapse
|
56
|
Gong YX. Research progress of effective components of traditional Chinese medicine in intervening apoptosis of renal tubular epithelial cells in diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119874. [PMID: 40280372 DOI: 10.1016/j.jep.2025.119874] [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: 12/11/2024] [Revised: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Apoptosis of renal tubular epithelial cells (RTECs) is a critical pathological feature of diabetic kidney disease (DKD), a primary contributor to end-stage renal disease (ESRD). Traditional Chinese medicine (TCM) has shown potential in modulating RTECs apoptosis and alleviating DKD progression, making it a promising area for further investigation. AIM OF THE STUDY This study aims to summarize the apoptotic pathways implicated in DKD, analyze existing research on the effects of TCM monomers and compounds on RTECs apoptosis, and elucidate the molecular mechanisms underlying these effects. Additionally, this study emphasizes the significant role of TCM in mitigating DKD progression. MATERIALS AND METHODS Relevant literature was systematically retrieved from ancient Chinese medicine texts and modern scientific databases, including CNKI, Web of Science, and PubMed, using keywords such as "Traditional Chinese Medicine", "Diabetic Kidney Disease", "Diabetic Nephropathy", "Renal Tubular Epithelial Cells", and "Apoptosis". The collected information was synthesized and analyzed. RESULTS This review systematically analyzed 187 relevant studies, focusing on the mechanisms and clinical applications of 16 TCM monomers and 20 TCM compounds in DKD treatment. Key bioactive compounds, such as berberine, astragaloside IV, and tanshinone IIA, have demonstrated renoprotective effects by mitigating oxidative stress and inflammation, as well as regulating critical signaling pathways, including PI3K/Akt, NF-κB, and TGF-β/Smad, to suppress RTECs apoptosis and decelerate DKD progression. Additionally, several TCM compounds have shown significant efficacy in clinical studies, reducing proteinuria and enhancing renal function, thereby reinforcing the therapeutic potential of TCM in DKD management. CONCLUSIONS RTECs apoptosis is a critical pathological feature of DKD. TCM exhibits significant therapeutic potential by intervening in this process through multiple pathways. This study highlights the ability of TCM to exert anti-apoptotic and renoprotective effects by modulating oxidative stress, inflammatory responses, and multiple cellular signaling pathways. The multi-component and multi-target characteristics of TCM offer a promising avenue for the development of novel therapeutic strategies. However, further rigorous research and high-quality clinical trials are required to validate its efficacy and elucidate its mechanisms of action.
Collapse
Affiliation(s)
- Yu Xin Gong
- Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
57
|
Talati YK, Dagar N, Gaikwad AB. Flavokawain A Attenuated Chronic Kidney Disease: Evidence From Network Pharmacology and Experimental Verification. Cell Biol Int 2025. [PMID: 40418741 DOI: 10.1002/cbin.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/16/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025]
Abstract
Chronic kidney disease (CKD) is a silent global epidemic affecting ~700 million people worldwide, contributing to rising mortality rates. Despite the variety of underlying causes, renal fibrosis is the key pathological feature of CKD. Flavokawain A (FKA), a natural chalcone, is thought to offer protective effects against CKD through its anti-inflammatory, antioxidant, and anti-fibrotic properties. This study aims to investigate the therapeutic potential of FKA against CKD, using network pharmacology (NP), molecular docking analysis, and In Vivo validation. GeneCards, SwissTargetPrediction, and SuperPred databases were utilized to identify therapeutic targets related to CKD and FKA. Protein-protein interactions (PPIs) were performed using the STRING database. Gene ontology and pathway enrichment analyses were performed with DAVID databases, followed by network construction in Cytoscape. For validation, molecular docking studies were performed using PyRx and tested at doses of 50 mg/kg and 100 mg/kg (p.o.) for 21 days using a unilateral ureteral obstruction (UUO) rat model. The study identified 109 therapeutic targets for FKA in relation to CKD, highlighting 11 hub targets and 78 potential pathways. Molecular docking showed strong binding efficacy with nuclear factor κB subunit 1 (NF-κB1) and matrix metallopeptidase 9 (MMP9). In vivo validation supported these findings, as FKA administration showed protective effects on kidney function and histology with the downregulation of extracellular matrix (ECM) markers, such as fibronectin (FN) and transforming growth factor β1 (TGF-β1), along with reduced expression of NF-κB1 and MMP9. These findings indicate that FKA could be a valuable therapeutic candidate for managing CKD by targeting NF-κB1 and MMP9.
Collapse
Affiliation(s)
| | - Neha Dagar
- Department of Pharmacy, Birla Institute of Technology and Science, Rajasthan, India
| | | |
Collapse
|
58
|
Gao L, Ma S, Yan S, Zhang H, Tian L, Li L, Diao L, Miao L, Yang X, Diao X. Pharmacokinetics, mass balance, and metabolism of [ 14C]FCN-437c, a selective and potent CDK4/6 inhibitor in humans. Cancer Chemother Pharmacol 2025; 95:57. [PMID: 40418395 DOI: 10.1007/s00280-025-04779-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/03/2025] [Indexed: 05/27/2025]
Abstract
This study investigated the pharmacokinetics, mass balance, and metabolism of [14C]FCN-437c, a selective and potent CDK4/6 inhibitor, in humans. Six healthy male Chinese subjects were administered a single oral dose of 200 mg [14C]FCN-437c (120 µCi), and plasma, urine, and feces samples were collected up to 456 h post-dose. The geometric mean Cmax of radioactivity in plasma and blood were 706 and 557 ng eq./mL, respectively, with a median Tmax of 5.0 h and a geometric mean t1/2 of 56.5 h in plasma. The primary route of elimination was fecal excretion, accounting for a mean of 77.16% of the dose, whereas urinary excretion constituted a mean of 19.19% of the administered radioactivity. UHPLC-HRMS analysis identified 12 metabolites in human plasma, urine, and feces, with 8 of them being phase I metabolites, and the major metabolic pathways were mono-oxidation and O-dealkylation. Additionally, 4 phase II metabolites were identified, including two glucuronides, one glutathione conjugate, and one cysteine conjugate. The study provides insights into the metabolic stability and clearance mechanisms of FCN-437c in human, which are essential for its further clinical development and dosing regimens.
Collapse
Affiliation(s)
- Lei Gao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Sheng Ma
- First Affiliated Hospital of Soochow University, Jiangsu, 215006, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Shu Yan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Hua Zhang
- First Affiliated Hospital of Soochow University, Jiangsu, 215006, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
| | - Ling Tian
- Liaoning Avanc Pharmaceutical Co., Ltd., Liaoning, 121013, China
| | - Lize Li
- Liaoning Avanc Pharmaceutical Co., Ltd., Liaoning, 121013, China
| | - Lei Diao
- Shanghai Fosun Pharmaceutical (Group) Co., Ltd, Shanghai, 200233, China
| | - Liyan Miao
- First Affiliated Hospital of Soochow University, Jiangsu, 215006, China.
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China.
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| | - Xiaoran Yang
- Liaoning Avanc Pharmaceutical Co., Ltd., Liaoning, 121013, China.
| | - Xingxing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China.
| |
Collapse
|
59
|
Hajnajafi K, Iqbal MA. Mass-spectrometry based metabolomics: an overview of workflows, strategies, data analysis and applications. Proteome Sci 2025; 23:5. [PMID: 40420110 PMCID: PMC12105183 DOI: 10.1186/s12953-025-00241-8] [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: 06/05/2024] [Accepted: 03/26/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Metabolomics, a burgeoning field within systems biology, focuses on the comprehensive study of small molecules present in biological systems. Mass spectrometry (MS) has emerged as a powerful tool for metabolomic analysis due to its high sensitivity, resolution, and ability to characterize a wide range of metabolites thus offering deep insights into the metabolic profiles of living systems. AIM OF REVIEW This review provides an overview of the methodologies, workflows, strategies, data analysis techniques, and applications associated with mass spectrometry-based metabolomics. KEY SCIENTIFIC CONCEPTS OF REVIEW We discuss workflows, key strategies, experimental procedures, data analysis techniques, and diverse applications of metabolomics in various research domains. Nuances of sample preparation, metabolite extraction, separation using chromatographic techniques, mass spectrometry analysis, and data processing are elaborated. Moreover, standards, quality controls, metabolite annotation, software for statistical and pathway analysis are also covered. In conclusion, this review aims to facilitate the understanding and adoption of mass spectrometry-based metabolomics by newcomers and researchers alike by providing a foundational understanding and insights into the current state and future directions of this dynamic field.
Collapse
Affiliation(s)
- Kosar Hajnajafi
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Mohammad Askandar Iqbal
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates.
- College of Medicine, Gulf Medical University, Ajman, United Arab Emirates.
| |
Collapse
|
60
|
Fan YJ, Fang JA, Li SF, Liu T, Liu WY, Hu YL, Wang RH, Li H, Sun DL, Zhang G, Zhang ZY. Colon Dialysis with Yishen Decoction Improves Autophagy Disorder in Intestinal Mucosal Epithelial Cells of Chronic Renal Failure by Regulating SIRT1 Pathway. Chin J Integr Med 2025:10.1007/s11655-025-3829-6. [PMID: 40418456 DOI: 10.1007/s11655-025-3829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2024] [Indexed: 05/27/2025]
Abstract
OBJECTIVE To explore the mechanism of colon dialysis with Yishen Decoction (YS) in improving the autophagy disorder of intestinal epithelial cells in chronic renal failure (CRF) in vivo and in vitro. METHODS Thirty male SD rats were randomly divided into normal, CRF, and colonic dialysis with YS groups by a random number table method (n=10). The CRF model was established by orally gavage of adenine 200 mg/(kg•d) for 4 weeks. CRF rats in the YS group were treated with colonic dialysis using YS 20 g/(kg•d) for 14 consecutive days. The serum creatinine (SCr) and urea nitrogen (BUN) levels were detected by enzyme-linked immunosorbent assay. Pathological changes of kidney and colon tissues were observed by hematoxylin and eosin staining. Autophagosome changes in colonic epithelial cells was observed with electron microscopy. In vitro experiments, human colon cancer epithelial cells (T84) were cultured and divided into normal, urea model (74U), YS colon dialysis, autophagy activator rapamycin (Ra), autophagy inhibitor 3-methyladenine (3-MA), and SIRT1 activator resveratrol (Re) groups. RT-PCR and Western blot were used to detect the mRNA and protein expressions of zonula occludens-1 (ZO-1), Claudin-1, silent information regulator sirtuin 1 (SIRT1), LC3, and Beclin-1 both in vitro and in vivo. RESULTS Colonic dialysis with YS decreased SCr and BUN levels in CRF rats (P<0.05), and alleviated the pathological changes of renal and colon tissues. Expressions of SIRT1, ZO-1, Claudin-1, Beclin-1, and LC3II/I were increased in the YS group compared with the CRF group in vivo (P<0.05). In in vitro study, compared with normal group, the expressions of SIRT1, ZO-1, and Claudin-1 were decreased, and expressions of Beclin-1, and LC3II/I were increased in the 74U group (P<0.05). Compared with the 74U group, expressions of SIRT1, ZO-1, and Claudin-1 were increased, whereas Beclin-1, and LC3II/I were decreased in the YS group (P<0.05). The treatment of 3-MA and rapamycin regulated autophagy and the expression of SIRT1. SIRT1 activator intervention up-regulated autophagy as well as the expressions of ZO-1 and Claudin-1 compared with the 74U group (P<0.05). CONCLUSION Colonic dialysis with YS could improve autophagy disorder and repair CRF intestinal mucosal barrier injury by regulating SIRT1 expression in intestinal epithelial cells.
Collapse
Affiliation(s)
- Yan-Jun Fan
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing-Ai Fang
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Su-Fen Li
- Department of Pathology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Ting Liu
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Wen-Yuan Liu
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ya-Ling Hu
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Rui-Hua Wang
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hui Li
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Da-Lin Sun
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Guang Zhang
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zi-Yuan Zhang
- Department of Nephrology, the First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
61
|
Zhang Y, Wu H, Jin M, Feng G, Wang S. The gut-heart axis: unveiling the roles of gut microbiota in cardiovascular diseases. Front Cardiovasc Med 2025; 12:1572948. [PMID: 40491716 PMCID: PMC12146390 DOI: 10.3389/fcvm.2025.1572948] [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: 02/13/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
The gut microbiome refers to the collective genomes of the approximately 1,000-1,150 microbial species found in the human gut, called the gut microbiota. Changing the gut microbiota composition has been shown to affect cardiovascular health significantly. Numerous studies have demonstrated the part that gut microbiota and its metabolites play in the development and course of several illnesses, including colon cancer, heart failure, stroke, hypertension, and inflammatory bowel disease. With cardiovascular diseases responsible for more than 31% of all fatalities globally, conditions like hypertension, atherosclerosis, and heart failure are serious global health issues. Developing preventive measures to fight cardiovascular diseases requires understanding how the gut microbiota interacts with the cardiovascular system. Understanding the distinctive gut microbiota linked to cardiovascular diseases has been made possible by microbial sequencing analysis. The gut microbiota and cardiovascular diseases are closely related, and more profound knowledge of this association may result in treatment strategies and broad guidelines for enhancing cardiovascular health through gut microbiome modification. This review summarizes the role of gut microbiota in cardiovascular diseases, highlighting their influence on disease progression and potential therapeutic implications.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, International Mongolian Hospital of Inner Mongolia, Hohhot, Inner Mongolia, China
| | - Huimin Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mu Jin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guirong Feng
- Department of Anesthesiology, International Mongolian Hospital of Inner Mongolia, Hohhot, Inner Mongolia, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Linzhi People’s Hospital, Linzhi, Tibet, China
| |
Collapse
|
62
|
Mohanty SK, Sahu VK, Singh BP, Suchiang K. Bidirectional upregulation of Klotho by triiodothyronine and baicalein: mitigating chronic kidney disease and associated complications in aged BALB/c mice. Biogerontology 2025; 26:114. [PMID: 40418372 DOI: 10.1007/s10522-025-10257-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
Chronic kidney disease (CKD) is a global health challenge marked by progressive renal decline and increased mortality. The interplay between CKD and hypothyroidism, particularly nonthyroidal low-triiodothyronine (T3) syndrome, exacerbates disease progression, driven by HPT axis dysfunction and reduced Klotho levels due to the Wnt/β-catenin pathway activation. This study explored Klotho as a link between CKD and hypothyroidism using an adenine-induced CKD aged mouse model. Exogenous T3 and baicalein (BAI), targeting the Wnt pathway, were used to upregulate Klotho expression. Combined T3 and BAI treatment significantly increased Klotho levels, surpassing individual effects, and suppressed key signaling molecules (TGF, NFκB, GSK3), mitigating renal fibrosis and CKD complications, including cardiovascular disorders and dyslipidemia. This bidirectional approach, enhancing Klotho via T3 and sustained Wnt pathway inhibition, offers a novel and effective strategy for CKD management, particularly in elderly patients with hypothyroidism.
Collapse
Affiliation(s)
- Saswat Kumar Mohanty
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India.
| | - Vikas Kumar Sahu
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India
| | - Bhanu Pratap Singh
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India
| | - Kitlangki Suchiang
- Department of Biochemistry, North Eastern Hill University, Shillong, 793022, India
| |
Collapse
|
63
|
Wu W, Tang W, Liang W, Li Q, Qi X, Gao R, Zhou P, Chen G, Xiong J, Gu X, Ding P, Yu M, Yang C, Wen S. GDF15 suppresses abdominal aortic aneurysm by upregulating AREG expression to adjust macrophage polarization. Int Immunopharmacol 2025; 159:114899. [PMID: 40414071 DOI: 10.1016/j.intimp.2025.114899] [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: 10/22/2024] [Revised: 01/19/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE Inflammation plays a key role in abdominal aortic aneurysm (AAA), with macrophages being crucial. Growth differentiation factor 15 (GDF15) is a new anti-inflammatory cytokine potentially useful in AAA diagnosis and treatment, but its role is unclear. METHODS AND RESULTS In mice with AAA, GDF15 expression was higher in lesioned tissues. Daily intraperitoneal injection of recombinant GDF15 (rGDF15) reduced aortic dilation, inflammation, degradation of aortic wall elastin and collagen, cellular apoptosis, and increased smooth muscle cells. GDF15 knockdown worsened AAA severity. Immunohistochemistry and immunofluorescence showed rGDF15 treatment reduced M1 macrophage polarization and enhanced M2 polarization, decreasing the M1/M2 ratio. GDF15 knockdown had the opposite effect. Additionally, Amphiregulin (AREG) expression increased with rGDF15 treatment and decreased with GDF15 knockdown. Immunofluorescence colocalization revealed lower AREG expression in M1 macrophages and higher AREG expression in M2 macrophages, suggesting that AREG may be involved in the regulation of macrophage polarization by GDF15 in AAA. Mechanistically, GDF15 upregulates AREG expression by activating the TGF-βR/SMAD2/3 signaling pathway, thereby inhibiting M1 polarization and promoting M2 polarization of macrophages. CONCLUSION This study demonstrates that exogenous injection of rGDF15 upregulates AREG expression and regulates macrophage polarization, thereby inhibiting AAA. GDF15 may not only serve as a diagnostic and prognostic marker for AAA but also as a potential molecular target for therapeutic intervention in AAA.
Collapse
Affiliation(s)
- Wanying Wu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenjing Tang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoyu Qi
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gezheng Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinrong Xiong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoying Gu
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiwu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Shuang Wen
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
64
|
Yu HR, Tiao MM, Huang SC, Sheu JJC, Tain YL, Sheen JM, Lin IC, Tsai CC, Huang LT, Hsu CN, Tsai CM, Lin YH, Lee PF, Su YT. Impact of maternal microplastic exposure on offspring lung structure and function: Insights into transcriptional misregulation and the TGF-β/α-SMA pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118397. [PMID: 40412254 DOI: 10.1016/j.ecoenv.2025.118397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/16/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
The "Developmental Origins of Health and Disease" (DOHaD) theory suggests that prenatal exposure to harmful environmental factors may impair fetal tissue development, increasing the risk of diseases later in life. This study investigated the effects of prenatal exposure to polystyrene microplastics (PS-MPs) on offspring lung development. Pregnant Sprague-Dawley rats were randomly assigned to receive PS-MPs in drinking water until delivery, with a control group receiving standard water. Offspring were assessed at 7 and 120 d after birth without further PS-MPs exposure. Histopathological examination at 7 d revealed PS-MPs deposits, alveolar collapse, and inflammation in lung tissue. Gene expression analysis showed disruptions in tight junctions, transcriptional regulation, and transforming growth factor-beta (TGF-β) pathways. By day 120, lung dysfunction and structural changes, consistent with emphysema were observed. These findings demonstrate that prenatal PS-MPs exposure adversely affects lung development potentially increasing the risk of respiratory diseases. Public health measures should address the potential hazards of microplastics to fetal health.
Collapse
Affiliation(s)
- Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
| | - Shun-Chen Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80761, Taiwan; School of Chinese Medicine, China Medical University, Taichung 40454, Taiwan.
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Kaohsiung 83301, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan; Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chih-Min Tsai
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan & College of Medicine, Chang Gung University, Taoyuan 330, Taiwan
| | - Yu-Hsiu Lin
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Pei-Fen Lee
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yu-Tsun Su
- Department of Pediatrics, E-Da Hospital, I-Shou University and School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
65
|
Lin H, Shao X, Gu H, Yu X, He L, Zhou J, Zhong Z, Guo S, Li D, Chen F, Song Y, Xu L, Wang P, Meng L, Chi J, Lian J. Akkermansia muciniphila ameliorates doxorubicin-induced cardiotoxicity by regulating PPARα-dependent mitochondrial biogenesis. NPJ Biofilms Microbiomes 2025; 11:86. [PMID: 40410194 PMCID: PMC12102390 DOI: 10.1038/s41522-025-00712-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/26/2025] [Indexed: 05/25/2025] Open
Abstract
Doxorubicin (DOX) is a key chemotherapeutic agent but is also a leading cause of DOX-induced cardiotoxicity (DIC), limiting its clinical use. Akkermansia muciniphila (A. muciniphila), known for its benefits as a probiotic in treating metabolic syndrome, has uncertain effects in the context of DIC. Here, 16S rRNA sequencing of fecal samples from anthracycline-treated patients and DIC mice revealed marked depletion of A. muciniphila. Cardiac transcriptomics, supported by in vitro experiments, showed that A. muciniphila colonization improved mitochondrial function and alleviated DIC by activating the PPARα/PGC1α signaling pathway in both normal and antibiotic-treated C57BL/6 mice. Further analysis uncovered a restructured microbiome-metabolome network following A. muciniphila administration, which contributed to DIC protection. Notably, A. muciniphila supplementation increased serum levels of the tryptophan metabolite indole-3-propionic acid (IPA), which binds to the cardiac aryl hydrocarbon receptor (AhR), leading to the activation of the PPARα/PGC1α signaling pathway. In conclusion, our study sheds light on the potential of A. muciniphila as a probiotic in mitigating DIC.
Collapse
Affiliation(s)
- Hui Lin
- Department of Cardiology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Xian Shao
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, China
| | - Haodi Gu
- College of Medicine, Shaoxing University, Shaoxing, China
| | - Xinrou Yu
- Department of Cardiology, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China
| | - Lingyan He
- Department of Traditional Chinese Medicine, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, China
| | - Jiedong Zhou
- Cardiac Arrhythmia Center, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zuoquan Zhong
- Department of Respiratory Medicine, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Shitian Guo
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Li
- Department of Haematology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Fei Chen
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Yongfei Song
- Department of Cardiology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Lili Xu
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, China
| | - Ping Wang
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Liping Meng
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China.
| | - Jufang Chi
- Department of Cardiology, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, China.
| | - Jiangfang Lian
- Department of Cardiology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
66
|
Lian K, Fan Q, Sheng S, Zhang K, Sun X, Kan C, Pan R, Guo Z. Metabolic Dysfunction-Associated Steatotic Liver Disease and Chronic Kidney Disease: Unraveling Connections and Advancing Therapies. BRATISL MED J 2025. [DOI: 10.1007/s44411-025-00189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 06/02/2025]
|
67
|
Chen W, Li ML, Zeng G, Xu XY, Yin SH, Xu C, Li L, Wen K, Yu XH, Wang G. Gut microbiota-derived metabolite phenylacetylglutamine in cardiovascular and metabolic disease. Pharmacol Res 2025:107794. [PMID: 40409519 DOI: 10.1016/j.phrs.2025.107794] [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: 03/17/2025] [Revised: 05/18/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
The aging of population coupled with unhealthy dietary habits among residents has led to a rise in the incidence of cardiovascular and metabolic diseases (CVMDs). Extensive research has highlighted the role of gut microbiota-derived metabolites in CVMDs. Among these metabolites, phenylacetylglutamine (PAGln), a meta-organismal prothrombotic metabolite, has been proved to promote the progression of CVMDs. This bacterial derived metabolite is a byproduct of amino acid comes from phenylalanine (Phe) in the diet. There are increasing evidence showing that the level of PAGln is associated with the risk of developing CVMDs. To provide a comprehensive understanding of the role of PAGln in CVMDs, this review delves into the production and metabolic pathways of PAGln and discusses the links of PAGln and the pathogenesis of CVMDs.
Collapse
Affiliation(s)
- Wan Chen
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China; Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, China
| | - Mei-Ling Li
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Guang Zeng
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Xiang-Yu Xu
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Shan-Hui Yin
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Can Xu
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, China
| | - Kaikai Wen
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 101400, China.
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, China.
| | - Gang Wang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical school, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
68
|
Guo R, Chen MN, Lin QH, Qi HM, Wang XQ, Li BY, Wang S, Xu SJ, Zhang Y, Liu W. LARS1 Promotes Tubular Epithelial Cells Epithelial Mesenchymal Transition in Chronic Kidney Disease by Inhibiting Lipophagy. Inflammation 2025:10.1007/s10753-025-02313-5. [PMID: 40397353 DOI: 10.1007/s10753-025-02313-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025]
Abstract
Tubulointerstitial fibrosis (TIF), a critical pathological hallmark in progressive chronic kidney disease (CKD), may be potentiated by renal lipid metabolism dysregulation and ectopic lipid deposition, though these processes likely exhibit bidirectional interactions with fibrotic progression Lipophagy is a type of selective autophagy that specifically recognizes lipid droplets and is accountable for lipid stability and metabolism. It serves as a link between lipid metabolism and autophagy. It was found that a positive correlation between elevated LARS1 expression and the severity of renal interstitial fibrosis in CKD patients. In Lars1+/- mice, we observed that the absence of LARS1 significantly reduced lipid deposition and TIF. Mechanistically, stimulation of HK-2 cells with TGF-β1 resulted in LARS1-mediated activation of mTORC1 and suppression of lipophagy, consequently leading to increased lipid accumulation and epithelial mesenchymal transition (EMT) through a defined mechanistic pathway. Collectively, our studies demonstrate that LARS1 plays a pivotal role in renal fibrosis at least in part by inhibiting lipophagy, suggesting that targeting LARS1 may represent a novel therapeutic strategy for patients with CKD.
Collapse
Affiliation(s)
- Rui Guo
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
- Department of Pathophysiology, Hebei North University, Zhangjiakou, 075000, China
| | - Mei-Ni Chen
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
| | - Qian-Hui Lin
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
| | - Hui-Min Qi
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
| | - Xiao-Qi Wang
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
| | - Bing-Yu Li
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
| | - Shuo Wang
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China
| | - Su-Juan Xu
- Department of Nephrology, Third Hospital of Hebei Medical University, Shijiazhuang, 050017, Hebei Province, China
| | - Yue Zhang
- Department of Diagnostics, Hebei Medical University, No. 361 Zhongshan East Rd, Shijiazhuang, 050017, Hebei Province, China.
| | - Wei Liu
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China.
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei Province, China.
- Hebei Key Laboratory of Forensic Medicine, Hebei Province, Shijiazhuang, 050017, China.
| |
Collapse
|
69
|
Zhi Y, Shu M, Tang P, Li Y, Guo M, Deng J, Mo H, Wu M, Liu B, Mai Y, Ling J, Zhao X, Zhang X, Zuo W. Overexpression of Decorin Optimizes the Treatment Efficacy of Umbilical Cord Mesenchymal Stem Cells in Bleomycin-Induced Pulmonary Fibrosis in Rats. Stem Cells Int 2025; 2025:6324980. [PMID: 40438789 PMCID: PMC12119169 DOI: 10.1155/sci/6324980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/02/2024] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a long-term, diffuse pulmonary parenchyma lesion that primarily affects middle-aged and older adults. It is characterized by pulmonary interstitial fibrosis of unknown cause. The death rate upon diagnosis is higher than that of many other cancer types. Mesenchymal stem cell (MSC) treatment of organ fibrosis is a hot topic in preclinical and clinical research because it effectively treats IPF. In recent years, decorin (DCN) has been regarded as a critical mediator for its anti-inflammatory and antifibrotic effects. The purpose of this study was to generate human umbilical cord MSCs (HUC-MSCs) that overexpress DCN and to investigate the safety, mechanism, and effectiveness of using these cells to cure pulmonary fibrosis caused by bleomycin (BLM). First, lentiviral (LV) particles carrying the therapeutic DCN gene (LV-DCN) and control LV particles were created and transfected using the plasmid vector GV208 to create a viral solution for infecting HUC-MSCs. These solutions were used to create a DCN overexpression cell line and an MSC-Con. cell line infected with the control lentivirus. Intratracheal injection of BLM was used to establish a rat model of pulmonary fibrosis. On the second day following modeling, different treatments were administered, and the body weight and survival status of the rats were noted. The relevant tests were performed on days 15 and 29 following modeling. The results demonstrated that the overexpression of DCN did not affect the properties of HUC-MSCs and that these cells were effective in treating IPF. MSC-Con. and MSC-DCN reduced systemic inflammation by reducing serum interleukin (IL) 1β. Both cell types successfully treated pulmonary fibrosis in rats, as demonstrated by hematoxylin and eosin (HE) and Masson staining. MSC-DCN showed better efficacy due to lower mortality, higher weight gain, less alveolar inflammation, and less fibrosis. The safety of venous transplantation with MSCs was established by HE staining of the heart, liver, spleen, and kidney, as well as serum lactate dehydrogenase (LDH), creatinine (CRE), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels. Immunohistochemical (IHC) staining of CD68 and CD206 in lung tissue and in vitro experiments on THP-1-induced M2 macrophage polarization and transforming growth factor-beta 1 (TGF-β1)-induced MRC-5 fibrosis indicated that MSC-DCN may mitigate lung inflammation by altering macrophage recruitment and polarization and inhibiting TGF-β1 expression to reduce fibrous hyperplasia and collagen deposition, thereby improving the treatment of BLM-induced IPF.
Collapse
Affiliation(s)
- Yaofeng Zhi
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Minghui Shu
- Department of Geriatric Medicine, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
| | - Pingsheng Tang
- Department of Cardiovascular, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
| | - Yingjie Li
- Department of Respiratory, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
| | - Min Guo
- Traditional Chinese Medicine Department, Guangdong Jiangmen Chinese Medicine College, Jiangmen, Guangdong, China
| | - Jiongrui Deng
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Haixin Mo
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Meimei Wu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Baoyi Liu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Yanyang Mai
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Jie Ling
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xulin Zhao
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wanli Zuo
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Department of Respiratory, The Affiliated Jiangmen Traditional Chinese Medicine Hospital of Jinan University, Jiangmen, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| |
Collapse
|
70
|
Cuevas-Delgado P, Miguel V, Lamas S, Barbas C, Rupérez FJ. Multiplatform Metabolomic Profiling of the Unilateral Ureteral Obstruction Murine Model of CKD. Int J Mol Sci 2025; 26:4933. [PMID: 40430073 PMCID: PMC12112560 DOI: 10.3390/ijms26104933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
In chronic kidney disease (CKD) research, animal models such as the unilateral ureteral obstruction (UUO) rodent model are crucial to understanding disease progression, particularly renal fibrosis. Despite its widespread use, the molecular mechanisms driving CKD remain incompletely understood. Given the interplay between metabolism and fibrosis, a comprehensive metabolomic analysis of UUO renal tissue is necessary. This study involved untargeted multiplatform analysis using liquid chromatography (LC), gas chromatography (GC), and capillary electrophoresis (CE) coupled with mass spectrometry (MS) to examine murine kidney tissue from the UUO model. The results highlight metabolic changes associated with tubulointerstitial fibrosis, which affect pathways such as the tricarboxylic acid (TCA) cycle, the urea cycle, and lipid metabolism. In particular, fibrosis impacts the lipidomic profile, with decreases in most lipid classes and increases in specific glycerophospholipids, hexosylceramides, and cholesterol esters. These findings demonstrate the value of a multiplatform approach in elucidating metabolic alterations in CKD, providing information on the underlying molecular mechanisms and paving the way for further research.
Collapse
Affiliation(s)
- Paula Cuevas-Delgado
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid 28660, Spain; (P.C.-D.); (C.B.)
| | - Verónica Miguel
- Program of Physiological and Pathological Processes, Centro de Biología Molecular “Severo Ochoa” (CBMSO, CSIC-UAM), c. Nicolás Cabrera 1, Madrid 28049, Spain; (V.M.); (S.L.)
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular “Severo Ochoa” (CBMSO, CSIC-UAM), c. Nicolás Cabrera 1, Madrid 28049, Spain; (V.M.); (S.L.)
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid 28660, Spain; (P.C.-D.); (C.B.)
| | - Francisco J. Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid 28660, Spain; (P.C.-D.); (C.B.)
| |
Collapse
|
71
|
Wang Y, Chen S, Zhou Z, Jiang J, Chen S. Tetrathiomolybdate alleviates bleomycin-induced pulmonary fibrosis by reducing copper concentration and suppressing EMT. Eur J Med Res 2025; 30:394. [PMID: 40390111 PMCID: PMC12087083 DOI: 10.1186/s40001-025-02640-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/27/2025] [Indexed: 05/21/2025] Open
Abstract
Pulmonary fibrosis (PF) is a disease characterized by dysregulated extracellular matrix deposition and aberrant fibroblast activation. Emerging evidence implicates that dysregulated copper metabolism contributed to fibrotic pathogenesis, yet its role and the therapeutic potential of copper modulation remain underexplored. This study investigated the involvement of cuproptosis, a programmed cell death induced by intracellular copper overload, in PF and evaluated the therapeutic efficacy of the copper chelator tetrathiomolybdate (TTM). In a bleomycin (BLM)-induced murine PF model, intratracheal BLM administration elevated lung copper levels, upregulated oligomerized DLAT, and exacerbated fibrosis, as evidenced by collagen deposition, α-smooth muscle actin, and transforming growth factor-beta expression. TTM treatment significantly attenuated fibrotic progression, reduced oxidative stress, and suppressed Olig-DLAT accumulation. In vitro, copper ionophores induced cuproptosis in bronchial epithelial cells, characterized by reduced viability, elevated intracellular Cu⁺, and Olig-DLAT aggregation, which were reversed by TTM. Furthermore, TTM mitigated TGF-β-driven epithelial-mesenchymal transition (EMT) and fibroblast-to-myofibroblast transition (FMT), downregulating collagen-1 and restoring E-cadherin expression. These findings establish cuproptosis as a novel mechanistic contributor to PF and highlight TTM's dual role in restoring copper homeostasis and inhibiting fibrogenic pathways, offering a promising therapeutic strategy for fibrotic lung diseases.
Collapse
Affiliation(s)
- Yajun Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Shuyang Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhou
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinjun Jiang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Shujing Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
72
|
Wang H, Chao L, Shen S, You P, Li L, Chen X, Hong Z, Chai Y. Exploring the pharmacological mechanism of Bu-Wang San on Alzheimer's disease through multiple GEO datasets of the human hippocampus, network pharmacology, and metabolomics based on GC-MS and UPLC-Q/TOF-MS. JOURNAL OF ETHNOPHARMACOLOGY 2025; 350:119994. [PMID: 40389089 DOI: 10.1016/j.jep.2025.119994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bu-Wang San (BWS) is a prominent traditional Chinese medicine known for calming the mind and promoting intelligence. It has been reported to improve learning and memory, enhance memory ability, and promote synaptic plasticity. However, the complexity of the material basis and the diversity of therapeutic targets of BWS on Alzheimer's disease (AD) have not been elucidated. AIM OF THE STUDY This study aimed to investigate the therapeutic material basis and the mechanism of BWS in AD treatment by comprehensively analyzing multiple GEO datasets of the human hippocampus, network pharmacology, and multi-platform metabolomics validation. MATERIALS AND METHODS Three GEO datasets of the human hippocampus were utilized to identify AD-associated targets using weighted gene co-expression network analysis (WGCNA) and differential analysis. Network pharmacology analyses were performed to investigate BWS's therapeutic material basis and predict the therapeutic targets of BWS on AD. A rat model was induced through the concurrent administration of AlCl3 and D-galactose to validate BWS's therapeutic potential and underlying mechanisms in AD. To validate the results of GEO data mining and network pharmacology, a comprehensive metabolomics approach integrating gas chromatography-mass spectrometry (GC-MS) and ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF/MS) was conducted on rat serum samples to uncover potential metabolic alterations and their associated pathways. RESULTS A total of 6367 genes were selected as AD drug targets through WGCNA analysis and enrichment analysis of disease-associated gene expression profiles in the GEO database. Network pharmacology was performed in this study for the identification of potential interactions between the components of BWS and its targets, TP53, STAT3, EGFR, MAOA, NOS3, PPARG, PRKCA, MAPK8, AChE, ARG1, among others, which were among the top 25 highest probable targets of BWS acting on AD. The multi-platform metabolomics indicated that amino sugar and nucleotide sugar metabolism, glycine, serine and threonine metabolism pathways, and other pathways may be associated with the AD model based on AlCl3 and D-galactose. The comparison of differential metabolites between the AD model group and the BWS intervention group revealed that 66 of the 97 differential metabolites exhibited a pullback trend, indicating a potential therapeutic effect of BWS on these metabolites. CONCLUSION This study builds a systematic strategy combining GEO datasets, network pharmacology, and multi-platform metabolomics and provides valuable insights into the pharmacological mechanism of BWS on AD. The results suggest that BWS may exert its therapeutic effects on AD by modulating the amino sugar and nucleotide sugar metabolism, glycerophospholipid metabolism, glycine, serine and threonine metabolism pathway and acting on the drug targets of ARG1, MAOA, AChE, XDH, GAD2 et al. This strategy provides a deep understanding of the molecular mechanisms of herbal medicine in treating AD at a systematic level.
Collapse
Affiliation(s)
- Hui Wang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Liang Chao
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Shuqi Shen
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Piaoxue You
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350108, China
| | - Ling Li
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Xiaofei Chen
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Zhanying Hong
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Yifeng Chai
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
73
|
Sun X, Ren Q, Liu X, Tan H, Zhan Z, Lin E, Long Y, Hong X, Zhou L, Liu Y. Matrix metalloproteinase-10 promotes kidney fibrosis by transactivating β-catenin signaling. Cell Death Discov 2025; 11:241. [PMID: 40382334 DOI: 10.1038/s41420-025-02521-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025] Open
Abstract
Kidney fibrosis is characterized by excessive accumulation of extracellular matrix (ECM) and serves as a hallmark of chronic kidney disease (CKD). The turnover of ECM is controlled by a family of matrix metalloproteinases (MMPs), endopeptidases that play a crucial role in ECM remodeling and other cellular processes. In this study, we demonstrate that MMP-10 was upregulated in a variety of animal models of kidney fibrosis and human kidney biopsies from CKD patients. Bioinformatics analyses and experimental validation reveal that MMP-10 activated β-catenin in a Wnt-independent fashion. Knockdown of endogenous MMP-10 expression in vivo inhibited β-catenin activation and ameliorated kidney injury and fibrotic lesions, whereas over-expression of exogenous MMP-10 aggravated β-catenin activation and kidney fibrosis after injury. We found that MMP-10 cleaved and activated heparin-binding EGF-like growth factor (HB-EGF) via ectodomain shedding, leading to EGF receptor (EGFR) tyrosine phosphorylation and β-catenin transactivation via a cascade of events involving extracellular signal-regulated kinases and glycogen synthase kinase-3β. Consistently, treatment with erlotinib, a small-molecule EGFR inhibitor, effectively mitigated MMP-10-mediated kidney injury and fibrotic lesions in a dose-dependent fashion. Furthermore, β-catenin activation reciprocally upregulated the expression of MMP-10, thereby perpetuating kidney damage by forming a vicious cycle. Collectively, these results underscore that MMP-10 promotes kidney fibrosis through EGFR-mediated transactivating β-catenin in a Wnt-independent fashion. Our findings suggest that targeting MMP-10 could be a novel strategy for treatment of fibrotic CKD.
Collapse
Affiliation(s)
- Xiaoli Sun
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Qian Ren
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Xi Liu
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Huishi Tan
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Zhanji Zhan
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Enqing Lin
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Yinyi Long
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Xue Hong
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China.
| | - Youhua Liu
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Renal Failure Research, Guangdong Provincial Institute of Nephrology, Guangzhou, China.
| |
Collapse
|
74
|
Wang S, Li Z, Wang X, Zhou J, Meng S, Zhuang J, Zhou Y, Zhao Q, Zhu C, Zhang Y, Shen S. Bioinformatics analysis of comorbid mechanisms between ischemic stroke and end stage renal disease. Sci Rep 2025; 15:17060. [PMID: 40379713 PMCID: PMC12084348 DOI: 10.1038/s41598-025-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
Ischemic stroke (IS) is a leading global cause of mortality and disability, particularly prominent in patients with end-stage renal disease (ESRD). Despite clinical evidence of their comorbidity, the molecular mechanisms underlying their interaction remain elusive. This study aims to identify shared biomarkers, gene regulatory networks, and therapeutic targets through integrative bioinformatics analyses. Gene expression datasets for IS (GSE16561, GSE22255) and ESRD (GSE37171, GSE142153) were obtained from gene expression omnibus (GEO). Weighted gene co-expression network analysis (WGCNA) and differential expression genes (DEGs) analysis identified shared genes and enriched pathways. Protein-protein interaction networks were constructed using STRING with clustering algorithms. Immune cell infiltration analysis was performed via CIBERSORT. Transcriptional regulatory networks were predicted using RcisTarget and miRcode. Key gene expressions were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in clinical samples. We identified 417 intersecting genes through WGCNA and 1712 shared differentially expressed genes. Among these, seven key genes (MRPL49, MRPS2, MRPS9, MRPS10, MRPS11, MRPS27, TFB1M) demonstrated central roles in mitochondrial function. Immune infiltration analysis revealed significant correlations with T cells and neutrophils. Pathway enrichment implicated these genes in transforming growth factor beta (TGF-β) signaling, p53 pathway, and G2/M checkpoint. Clinical validation confirmed significant downregulation of MRPS9, MRPS10, MRPS11, MRPS27 and TFB1M in comorbid patients. This study systematically elucidates the mitochondrial-immune interaction mechanisms in IS-ESRD comorbidity, highlighting the pivotal role of mitochondrial ribosomal protein (MRP) family genes in regulating cellular energetics and inflammatory responses. These findings provide new foundations for targeted therapies.
Collapse
Affiliation(s)
- Shuhong Wang
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, Guangdong, China
| | - Zhongda Li
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Xiao Wang
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Jiexue Zhou
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Shandong Meng
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Jinyang Zhuang
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Yan Zhou
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Qin Zhao
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Chunli Zhu
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China
| | - Yusheng Zhang
- Department of Neurology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, Guangdong, China.
| | - Sheng Shen
- Department of Organ Transplantation, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, 510317, Guangdong, China.
| |
Collapse
|
75
|
Pei T, Li W, Zhou Z, Zhang Q, Yu G, Yin S, Chen H, Tang J. The relationship between tryptophan metabolism and gut microbiota: Interaction mechanism and potential effects in infection treatment. Microbiol Res 2025; 298:128211. [PMID: 40393170 DOI: 10.1016/j.micres.2025.128211] [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: 02/19/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Human health is influenced by the gut microbiota, particularly in aspects of host immune homeostasis and intestinal immune response. Tryptophan (Trp) not only acts as a nutrient enhancer but also plays a critical role in the balance between host immune tolerance and gut microbiota maintenance. Both endogenous and bacterial metabolites of Trp, exert significant effects on gut microbial composition, microbial metabolism, the host immunity and the host-microbiome interface. Trp metabolites regulate host immunity by activating aryl hydrocarbon receptor (AhR), thereby contributing to immune homeostasis. Among Trp metabolites, AhR ligands include endogenous metabolites (such as kynurenine), and bacterial metabolites (such as indole and its derivatives). Here, we present a comprehensive analysis of the relationships between Trp metabolism and 14 key microbiota, encompassing fungi (e.g., Candida albicans, Aspergillus), bacteria (e.g., Ruminococcus gnavus, Bacteroides, Prevotella copri, Clostridium difficile, Escherichia coli, lactobacilli, Mycobacterium tuberculosis, Pseudomonas aeruginosa, Staphylococcus aureus, Helicobacter. Pylori), and viruses (e.g., SARS-CoV-2, influenza virus). This review clarifies how the gut microbiota regulates Trp metabolism and uncovers the underlying mechanisms of these interactions. And increased mechanistic insight into how the microbiota modulate the host immune system through Trp metabolism may allow for the identification of innovative therapies that are specifically designed to target Trp absorption, Trp metabolites, the gut microbiota, or interactions between Trp and gut microbiota to treat both intestinal and extra-intestinal inflammation as well as microbial infections.
Collapse
Affiliation(s)
- Tongchao Pei
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Wenweiran Li
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Ziyang Zhou
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Qinyu Zhang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Guohong Yu
- Department of Emergency Medicine, Baoshan Second People's Hospital, Baoshan College of Traditional Chinese Medicine, Baoshan 678000, China
| | - Sokun Yin
- Department of Emergency Medicine, Luoping County People's Hospital, Qujing 655800, China
| | - Hui Chen
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China.
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China.
| |
Collapse
|
76
|
Chen Y, Zhang J, Feng X, Ma Q, Sun C. Single-cell RNA-seq uncovers lineage-specific regulatory alterations of fibroblasts and endothelial cells in ligamentum flavum hypertrophy. Front Immunol 2025; 16:1569296. [PMID: 40443657 PMCID: PMC12119296 DOI: 10.3389/fimmu.2025.1569296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/21/2025] [Indexed: 06/02/2025] Open
Abstract
Background Lumbar spinal stenosis (LSS) represents a major global healthcare burden resulting in back pain and disorders of the limbs among the elderly population. The hypertrophy of ligamentum flavum (HLF), marked by fibrosis and inflammation, significantly contributes to LSS. Fibroblasts and endothelial cells are two important cells in the pathological process of ligamentum flavum (LF) fibrosis and inflammation. These two cells exhibit heterogeneity in various fibrotic diseases, yet their heterogeneity in LF fibrosis remains poorly defined. Methods Using single-cell RNA-seq, we examined the alterations of fibroblasts, endothelial cells, and key genes in the hypertrophic LF, aiming to establish a comprehensive single-cell atlas of LF to identify high-priority targets for pharmaceutical treatment of LSS. Results Here, we find there are five distinct subpopulations of LF fibroblasts: secretory-papillary, secretory-reticular, mesenchymal, pro-inflammatory, and unknown. Importantly, in HLF, the proportion of mesenchymal fibroblast subpopulations increases significantly compared to normal LF (NLF), reflecting their close association with the pathogenesis of HLF. Furthermore, critical target genes that might be involved in HLF and fibrosis, such as MGP, ASPN, OGN, LUM, and CTSK, are identified. In addition, we also investigate the heterogeneity of endothelial cells and highlight the critical role of AECs subpopulation in LF fibrosis. Conclusion This study will contribute to our understanding of the pathogenesis of HLF and offer possible targets for the treatment of fibrotic diseases.
Collapse
Affiliation(s)
| | | | | | - Qinghong Ma
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chao Sun
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
77
|
Sun Q, Xie L, An H, Chen W, Yang Q, Wang P, Tang Y, Peng C. Characterizing hub biomarkers for metabolic-induced endothelial dysfunction and unveiling their regulatory roles in EndMT through RNA sequencing and machine learning approaches. Front Cardiovasc Med 2025; 12:1585030. [PMID: 40443971 PMCID: PMC12119472 DOI: 10.3389/fcvm.2025.1585030] [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: 02/28/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Background Metabolic disorder and endothelial dysfunction (ED) are key events in the development and pathophysiology of atherosclerosis and are associated with an elevated risk of Cardiovascular disease (CVD). The pathophysiology remains incompletely understood. Methods Leftover serum samples were collected and stored at -20 °C until study. Serum specimens were mixed to obtain pooled high glucose serum (GLU group) (11.97 ± 2.09 mmol/L); pooled elevated low-density lipoprotein serum (LDL group) [3.465 (3.3275, 3.6425 mmol/L)]; pooled high triglycerides serum (1.15 ± 0.35 mmol/L) (TG group); Subsequently, Human umbilical vein endothelial cells (HUVECs) were exposed to culture media supplemented with these pooled serum or control serum for 72 h. Whole transcriptome sequencing was performed to characterize gene expression profiles and data were analyzed using GSEA, GO, KEGG. qPCR was used to validate the gene expression. Results A total of 306 mRNAs and 523 lncRNAs were identified as differentially expressed in the GLU group, 335 mRNAs and 471 lncRNAs in the LDL group, and 364 mRNAs and 562 lncRNAs in the TG group, compared to the control group. These genes are primarily involved in inflammation, lipid metabolism, and EndMT pathways. By integrating differentially expressed mRNA and curated EndMT-related gene sets from the KEGG, GO, and dbEMT2.0 databases, we identified 52 differentially expressed genes associated with EndMT under metabolic stress conditions. Utilizing machine learning techniques, we established an EndMT-associated gene diagnostic signature comprising CD36, ISG15, HSPB2, and IRS2 for the diagnosis of AS, which achieved an AUC of 0.997. The model was subsequently validated across three independent external cohorts (GSE43292, GSE28829, GSE163154), in which it consistently demonstrated strong diagnostic performance, with AUC values of 0.958, 0.808, and 0.884, respectively. The ceRNA networks associated with EndMT are constructed and related lncRNAs including LINC002381, VIM-AS1, and ELF-AS1 were significantly upregulated in peripheral blood samples. Conclusions This study identified novel biomarkers for ED. These findings may provide both a potential biomarker and therapeutic target for the prevention and treatment of atherosclerosis and CAD.
Collapse
Affiliation(s)
- Qi Sun
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Longchuan Xie
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - He An
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wei Chen
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qirong Yang
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Peng Wang
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yijun Tang
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Chunyan Peng
- Clinical Molecular Diagnostic Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
78
|
Fu Y, Land M, Kavlashvili T, Cui R, Kim M, DeBitetto E, Lieber T, Ryu KW, Choi E, Masilionis I, Saha R, Takizawa M, Baker D, Tigano M, Lareau CA, Reznik E, Sharma R, Chaligne R, Thompson CB, Pe'er D, Sfeir A. Engineering mtDNA deletions by reconstituting end joining in human mitochondria. Cell 2025; 188:2778-2793.e21. [PMID: 40068680 DOI: 10.1016/j.cell.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/22/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025]
Abstract
Recent breakthroughs in the genetic manipulation of mitochondrial DNA (mtDNA) have enabled precise base substitutions and the efficient elimination of genomes carrying pathogenic mutations. However, reconstituting mtDNA deletions linked to mitochondrial myopathies remains challenging. Here, we engineered mtDNA deletions in human cells by co-expressing end-joining (EJ) machinery and targeted endonucleases. Using mitochondrial EJ (mito-EJ) and mito-ScaI, we generated a panel of clonal cell lines harboring a ∼3.5 kb mtDNA deletion across the full spectrum of heteroplasmy. Investigating these cells revealed a critical threshold of ∼75% deleted genomes, beyond which oxidative phosphorylation (OXPHOS) protein depletion, metabolic disruption, and impaired growth in galactose-containing media were observed. Single-cell multiomic profiling identified two distinct nuclear gene deregulation responses: one triggered at the deletion threshold and another progressively responding to heteroplasmy. Ultimately, we show that our method enables the modeling of disease-associated mtDNA deletions across cell types and could inform the development of targeted therapies.
Collapse
Affiliation(s)
- Yi Fu
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Max Land
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tamar Kavlashvili
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruobing Cui
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Minsoo Kim
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily DeBitetto
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Toby Lieber
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Keun Woo Ryu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elim Choi
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ignas Masilionis
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rahul Saha
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meril Takizawa
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daphne Baker
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marco Tigano
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Caleb A Lareau
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roshan Sharma
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronan Chaligne
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Howard Hughes Medical Institute, New York, NY, USA
| | - Agnel Sfeir
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
79
|
Zhang ZY, Wang YM, Wang N, Wang YS, Zhang H, Wang D, Wang LX, Cui HT, Wen WB, Lv SQ, Cao YJ. Shenzhuo formulation ameliorates diabetic nephropathy by regulating cytochrome P450-mediated arachidonic acid metabolism. World J Diabetes 2025; 16:103511. [DOI: 10.4239/wjd.v16.i5.103511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/31/2024] [Accepted: 02/17/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a major complication of diabetes, marked by progressive renal damage and an inflammatory response. Although research has investigated the pathological mechanisms underlying DN, effective treatment options remain limited.
AIM To evaluate the therapeutic impact of Shenzhuo formulation (SZF) on a DN mouse model and to examine its potential molecular mechanisms using transcriptomic and metabolomic approaches.
METHODS We established a DN mouse model through a high-fat diet combined with streptozotocin (STZ) injection, followed by SZF treatment. We analyzed SZF’s effects on gene expression and metabolite profiles in renal tissues of DN mice using transcriptomics and metabolomics techniques. Additionally, based on transcriptomic and non-targeted metabolomic findings, we further assessed SZF’s influence on the expression of factors related to the cytochrome P450 (CYP450)-mediated arachidonic acid (AA) metabolism pathway, as well as its effects on inflammation and oxidative stress.
RESULTS SZF intervention significantly decreased hyperglycemia and mitigated renal function impairment in DN mice. Pathological analysis revealed that SZF treatment improved renal tissue damage, reduced fibrosis, and diminished glycogen deposition. Transcriptomic analysis indicated that SZF influenced mRNA expression of CYP450-related genes, including Cyp2j13, Cyp2b9, Pla2 g2e/Cyp4a12a, Cyp4a32, Cyp2e1, and Cyp4a14. Non-targeted metabolomic results demonstrated that SZF altered the levels of metabolites associated with the AA metabolic pathway, including 5,6-EET, 14,15-EET, phosphatidylcholine, and 20-HETE. Further experiments showed that SZF upregulated the expression of CYP4A and CYP2E proteins in renal tissue, as well as CYP2J and CYP2B proteins. Additionally, SZF significantly reduced the expression of inflammatory factors in renal tissue, enhanced antioxidant enzyme activity, and alleviated oxidative stress.
CONCLUSION SZF exerts anti-inflammatory and antioxidant effects by regulating CYP450-mediated AA metabolism, leading to improved renal function and improved pathological state in DN mice.
Collapse
Affiliation(s)
- Zhong-Yong Zhang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Yu-Ming Wang
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ning Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Yuan-Song Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Hui Zhang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Duo Wang
- North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Li-Xin Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Huan-Tian Cui
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Wei-Bo Wen
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Shu-Quan Lv
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou 061012, Hebei Province, China
| | - Yong-Jun Cao
- Department of Endocrinology, Nantong Affiliated Hospital, Nanjing University of Traditional Chinese Medicine, Nantong 226000, Jiangsu Province, China
| |
Collapse
|
80
|
Koval A, Mamadalieva NZ, Mamadalieva R, Jalilov F, Katanaev VL. Success and Controversy of Natural Products as Therapeutic Modulators of Wnt Signaling and Its Interplay with Oxidative Stress: Comprehensive Review Across Compound Classes and Experimental Systems. Antioxidants (Basel) 2025; 14:591. [PMID: 40427472 PMCID: PMC12108534 DOI: 10.3390/antiox14050591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The highly conserved Wnt signaling pathway, a complex network critical for embryonic development and adult tissue homeostasis, regulates diverse cellular processes, ultimately influencing tissue organization and organogenesis; its dysregulation is implicated in numerous diseases, and it is known to be affected by oxidative pathways. This report reviews the recent literature on major classes of natural products with pronounced anti-oxidant properties, such as cardiac glycosides, steroid saponins, ecdysteroids, withanolides, cucurbitacins, triterpenes, flavonoids, and iridoids, that modulate its activity in various pathological conditions, summarizing and critically analyzing their effects on the Wnt pathway in various therapeutically relevant experimental models and highlighting the role of ROS-mediated crosstalk with Wnt signaling in these studies. Models reviewed include not only cancer but also stroke, ischemia, bone or kidney diseases, and regenerative medicine, such as re-epithelialization, cardiac maintenance, and hair loss. It highlights the paramount importance of modulating this signaling by natural products to define future research directions. We also discuss controversies identified in the mode of action of several compounds in different models and directions on how to further improve the quality and depth of such studies.
Collapse
Affiliation(s)
- Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
| | - Nilufar Z. Mamadalieva
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Faculty of Medicine, Alfraganus University, Yuqori Qoraqamish Str. 2a, Tashkent 100190, Uzbekistan;
- Institute of the Chemistry of Plant Substances, Uzbekistan Academy of Sciences, Mirzo Ulugbek Str. 77, Tashkent 100170, Uzbekistan
| | - Rano Mamadalieva
- Faculty of Medicine, Namangan University of Business and Science, Beshkapa Str. 111, Namangan 160100, Uzbekistan;
| | - Fazliddin Jalilov
- Faculty of Medicine, Alfraganus University, Yuqori Qoraqamish Str. 2a, Tashkent 100190, Uzbekistan;
| | - Vladimir L. Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Oncology Research Center, Qatar Biomedical Research Institute (QBRI), College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha P.O. Box 34110, Qatar
| |
Collapse
|
81
|
Bustani G, Alghetaa H, Mohammed A, Nagarkatti M, Nagarkatti P. The aryl hydrocarbon receptor: a new frontier in male reproductive system. Reprod Biol Endocrinol 2025; 23:70. [PMID: 40369574 PMCID: PMC12076877 DOI: 10.1186/s12958-025-01401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor historically recognized for its role in the regulation of toxicity mediated by environmental chemicals. Recent research points to AhR's critical participation in male reproductive physiology, particularly in spermatogenesis, hormone signaling, and the maintenance of sperm quality. Both endogenous ligands (e.g., dietary and gut microbiota-derived metabolites) and exogenous pollutants (e.g., dioxins and benzo-α-pyrene) influence AhR-mediated pathways, making it a key link between environmental exposures and male fertility. RESULTS This review highlights AhR's influence on the male reproductive system, emphasizing the role of endogenous AhR ligands and AhR expression in the maturation and function of male reproductive organs. Environmental AhR agonists have been shown to induce oxidative stress, hormonal imbalance, and sperm DNA damage, which impact harmfully on the spermatogenesis process, which leads to reproductive abnormalities. Conversely, certain natural compounds such as resveratrol, curcumin, and lycopene appear to antagonize AhR activation and reduce its negative effects, thus offering potential protective benefits against male reproductive toxicity. Nevertheless, discrepancies persist regarding the exact interplay between AhR signaling and critical reproductive hormones such as testosterone and LH, and it remains unclear how transgenerational epigenetic changes triggered by AhR activation might affect long-term male fertility. CONCLUSION AhR is pivotal in male reproductive physiology, influencing spermatogenesis, sperm quality, and hormone regulation through its interactions with both endogenous and environmental ligands. Persistent pollutants such as dioxins and polycyclic aromatic hydrocarbons cause oxidative damage and hormonal disturbances via AhR, contributing to reduced sperm quality and fertility.
Collapse
Affiliation(s)
- Ghadeer Bustani
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Abu Gharib Road, Al-Ameria Ave., Baghdad, Iraq
- The Islamic University, Najaf, Iraq
| | - Hasan Alghetaa
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Abu Gharib Road, Al-Ameria Ave., Baghdad, Iraq.
| | - Amira Mohammed
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Abu Gharib Road, Al-Ameria Ave., Baghdad, Iraq
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
82
|
Meng W, Li N, Lv F, Chen B, Lu S, Zhang J, Zhang T, Tao Q, Zhou Y, Ma L, Guan Y. SiRNA-Targeting TGF-β1 Based on Nanoparticle-Coated Ureteral Stents to Inhibit Ureteral Stricture. ACS Biomater Sci Eng 2025. [PMID: 40359485 DOI: 10.1021/acsbiomaterials.4c01925] [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: 05/15/2025]
Abstract
Ureteral stricture is a difficult urological problem with no optimal solution and is the result of scar hyperplasia and fibrosis caused by ureteral injury. Preventing the formation of ureteral strictures around drug-loaded ureteral stents is at the heart of the current research. TGF-β1 is a key factor affecting collagen deposition and fiber formation. Therefore, in this study, we established a rabbit ureteral stricture model, implanted a ureteral stent loaded with TGF-β1-siRNA for treatment, and compared the histopathology of ureteral stricture and the protein expression of genes related to the formation of stricture between different groups to test their therapeutic effects. We used sustained- and slow-release properties of the nanoparticles that were confirmed through in vitro experiments. The results of the fluorescence immunoassay showed that siRNA loaded by ureteral stents had high transfection efficiency on human ureter epithelial cells in vivo. In addition, the rabbit ureteral stricture model experiment verified that TGF-β1-siRNA could effectively transfect into ureteral tissues and inhibit the expression of TGF-β1, thereby inhibiting ureteral stricture. At the same time, the images of rabbit gross anatomy specimens showed that the hydronephrosis could also be effectively relieved. In summary, all the results mentioned above suggest that ureteral stents combined with RNA interference technology and a nanoparticle delivery system have broad prospects for clinical application in the suppression of ureteral stricture.
Collapse
Affiliation(s)
- Wei Meng
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Ningning Li
- Xinglin College, Nantong University, Nantong 226001, China
| | - Feng Lv
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Bo Chen
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Shuaijiang Lu
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jiayi Zhang
- Xinglin College, Nantong University, Nantong 226001, China
| | - Tong Zhang
- Xinglin College, Nantong University, Nantong 226001, China
| | - Qianyu Tao
- Xinglin College, Nantong University, Nantong 226001, China
| | - Youlang Zhou
- Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Limin Ma
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yangbo Guan
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
83
|
Ansari E, Kumar R, Ratnam A. Gold-NHC complexes: from synthetic aspects to anti-cancer activity. Dalton Trans 2025; 54:7553-7601. [PMID: 40171803 DOI: 10.1039/d5dt00118h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Recent advancements in Au(I)-N-heterocyclic carbene (NHC) complexes have demonstrated significant potential for developing novel anticancer agents. These complexes exhibit unique properties, such as a strong affinity for thiol and selenol-containing biomolecules, which enable the selective targeting of cancer cells while minimising effects on healthy tissues. Recent studies have explored various structural modifications to enhance the anticancer efficacy of Au(I)-NHC complexes, including ligand substitution, incorporation of bioactive moieties, and hybridisation with other metal complexes. Mechanistic investigations have revealed that these complexes induce apoptosis through multiple pathways, such as inhibition of thioredoxin reductase (TrxR), disruption of mitochondrial function, and generation of reactive oxygen species (ROS). The introduction of NHC ligands is particularly advantageous, as they provide stability and tunability to the Au(I) centre, allowing for the optimisation of pharmacokinetic and pharmacodynamic properties. Moreover, the emergence of Au(I)-NHC complexes with dual-action mechanisms, combining anticancer activity with antiangiogenic or anti-inflammatory properties, has further broadened their therapeutic potential. This review article highlights the most recent breakthroughs in the design, synthesis, and biological evaluation of Au(I)-NHC complexes, emphasizing their promise as a new class of targeted anticancer therapeutics. While primarily focused on Au(I) complexes, it also includes a brief discussion of selected Au(III) complexes for comparison.
Collapse
Affiliation(s)
- Eklakh Ansari
- Department of Chemistry, DDU Gorakhpur University, Gorakhpur-273009, India.
| | - Ravi Kumar
- Department of Chemistry, National Institute of Technology, Srinagar, 190006, India
| | - Anand Ratnam
- Department of Chemistry, DDU Gorakhpur University, Gorakhpur-273009, India.
| |
Collapse
|
84
|
Ouyang W, Deng Z, Li Y, Chi W, Huang Z, Zhan C, Li M, Wang D, Li F, Liu Y, Ling L. Traditional Chinese medicine in cerebral infarction: Integrative strategies and future directions. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156841. [PMID: 40393244 DOI: 10.1016/j.phymed.2025.156841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/09/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Cerebral infarction is a predominant cause of global disability and mortality, characterized by pathogenesis that includes vascular stenosis, thrombosis, ischemic necrosis, and neuroinflammation. Despite progress in medical science, effective treatments for cerebral infarction are still constrained, prompting the investigation of alternative therapeutic strategies. PURPOSE The objective of this review is to assess the efficacy of Traditional Chinese Medicine (TCM) as a treatment for cerebral infarction, emphasizing its mechanisms of action, effectiveness, and clinical relevance. STUDY DESIGN An extensive review of the existing literature regarding the role of TCM in the management of cerebral infarction, encompassing investigations on specific remedies, Chinese medicinal formulations, and contemporary advancements in preparation methodologies. METHODS This review analyzes diverse TCM remedies recognized for their antioxidant, anti-inflammatory, and neuroprotective properties. Furthermore, it examines the synergistic effects of Chinese medicine formulations in modulating inflammation, enhancing blood circulation, and facilitating neural repair. Contemporary technological advancements that improve the accuracy and efficacy of Chinese medicine are also taken into account. RESULTS TCM has shown considerable promise in tackling the complex aspects of cerebral infarction. Specific remedies and formulations have demonstrated potential in modulating inflammatory responses, enhancing cerebral blood flow, and promoting neural repair mechanisms. Contemporary formulations have enhanced these effects, facilitating more precise and effective treatment. CONCLUSION While TCM presents a promising multi-faceted and multi-tiered strategy for addressing cerebral infarction, obstacles such as elucidating mechanisms and achieving standardization must be addressed. Continued research and clinical trials are crucial to fully exploit the therapeutic potential of TCM in the management of cerebral infarction.
Collapse
Affiliation(s)
- Wenhao Ouyang
- Department of Neurology, Shenzhen Hospital, Southern Medical University, No.1333 Xinhu Road, Shenzhen, Guangdong 518000, China
| | - Zekang Deng
- School of Clinical Medicine, Nanchang Medical College, Nanchang, Jiangxi 330006, China
| | - Yudi Li
- Department of Neurology, Shenzhen Hospital, Southern Medical University, No.1333 Xinhu Road, Shenzhen, Guangdong 518000, China
| | - Wenjun Chi
- Department of Neurology, Shenzhen Hospital, Southern Medical University, No.1333 Xinhu Road, Shenzhen, Guangdong 518000, China
| | - Zhenjun Huang
- Department of Medical Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Chengyi Zhan
- School of Clinical Medicine, Nanchang Medical College, Nanchang, Jiangxi 330006, China
| | - Min Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Dongtao Wang
- Department of Traditional Chinese Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524037, China; Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China.
| | - Fangming Li
- Department of Neurology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, No. 1298, Xueyuan Avenue, Shenzhen, Guangdong 518000, China.
| | - Yajie Liu
- Department of Neurology, Shenzhen Hospital, Southern Medical University, No.1333 Xinhu Road, Shenzhen, Guangdong 518000, China.
| | - Li Ling
- Department of Neurology, Shenzhen Hospital, Southern Medical University, No.1333 Xinhu Road, Shenzhen, Guangdong 518000, China.
| |
Collapse
|
85
|
Gholizadeh N, Yousefian M, Mohammadpour H, Razavi AE, Talaei S, Sheykhbahaei N. Long non-coding RNAs PVT1, CCAT2, and TCF7L2, and miR-33 and c-Myc expression in oral squamous cell carcinoma and oral lichen planus patients. J Craniomaxillofac Surg 2025:S1010-5182(25)00136-2. [PMID: 40360347 DOI: 10.1016/j.jcms.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 03/09/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE The objective of this study was to assess the potential of TCF7L2, CCAT2, and PVT1 LncRNAs, c-Myc, and miR-33 as biomarkers for early diagnosis and differentiation of oral squamous cell carcinoma (OSCC) and premalignant lesions. DESIGN Bioinformatics tools, including COSMIC, GeneMANIA, PathVisio, KEGG Pathway Database, IntOGen, and WikiPathways, were used to investigate the signaling pathways of cancer-associated genes. The limma package was utilized for statistical analysis to identify Differentially Expressed Genes (DEGs) between OSCC tumor and normal samples. The regulatory microRNAs were analyzed using miRDB, miRWalk, and TargetScan. The type of cancer for analysis was selected using IntOGen. The expression levels of LncRNAs, miR-33, and c-Myc were measured by polymerase chain reaction (PCR) in 28 OLP and 30 OSCC tissue samples, compared to 30 healthy and 30 OSCC-adjacent tissue specimens as control groups. Data were analyzed using the Mann-Whitney test, receiver operating characteristic (ROC) curve, and multiple linear regression. RESULTS The expression of c-Myc (3.53 ± 2.78 vs 0.93 ± 0.50), PVT1 (10.94 ± 8.49 vs 0.91 ± 0.48), CCAT2 (11.77 ± 10.00 vs 0.92 ± 0.95), and TCF7L2 (6.48 ± 4.30 vs 1.27 ± 0.96) was significantly higher in OSCC samples compared to OLP (P < 0.001). Conversely, miR-33 expression was significantly lower in OSCC samples (0.24 ± 0.25 vs 4.90 ± 3.90). There was a significant correlation between c-Myc, CCAT2, PVT1, and miR-33 expression and clinicopathological characteristics of OSCC specimens. In OSCC samples, c-Myc, PVT1, CCAT2, and TCF7L2 showed a significant positive correlation with each other, while miR-33 expression was negatively correlated with the overexpression of other genes. The area under the curve (AUC) for c-Myc, PVT-1, CCAT2, miR-33, and TCF7L2 were 0.917, 1.000, 0.979, 0.006, and 0.929, respectively. CONCLUSIONS Our findings suggest that c-Myc and LncRNAs (TCF7L2, PVT1, and CCAT2) are upregulated and miR-33 is downregulated in OSCC compared to OLP samples. These genes may serve as potential genetic biomarkers for diagnosis and prediction of clinicopathological features of OSCC.
Collapse
Affiliation(s)
- Narges Gholizadeh
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran; Department of Oral & Maxillofacial Surgery, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Marzieh Yousefian
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Hadiseh Mohammadpour
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirnader Emami Razavi
- Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanam Talaei
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran
| | - Nafiseh Sheykhbahaei
- Department of Oral & Maxillofacial Medicine, School of Dentistry, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
86
|
Wu Y, Li T, Chen B, Sun Y, Song L, Wang Y, Bian Y, Qiu Y, Yang Z. Tryptophan Indole Derivatives: Key Players in Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2025; 18:1563-1574. [PMID: 40386349 PMCID: PMC12083488 DOI: 10.2147/dmso.s511068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 05/02/2025] [Indexed: 05/20/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex clinical syndrome characterized by insulin resistance and associated with abnormal amino acid metabolism. Tryptophan is an aromatic dietary amino acid that affects T2DM by regulating glycolipid metabolism and insulin resistance. When tryptophan reaches the intestine, it is converted by gut microbiota and tryptophanase into indole derivatives such as indoleacetic acid, indolepropionic acid, and indolealdehyde. These indole derivatives may enhance insulin sensitivity, stimulate insulin secretion, and exert functions such as lowering blood glucose, regulating hepatic oxidative stress, reducing intestinal inflammation, and improving islet cell morphology by acting on the aryl hydrocarbon receptor (AHR) or Pregnane X receptor (PXR). In summary, this review aims to examine the interactions between tryptophan indole derivatives and T2DM thoroughly, elucidate potential therapeutic approaches, and pinpoint areas for further research.
Collapse
Affiliation(s)
- Yingli Wu
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Tiangang Li
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Biaohua Chen
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Yi Sun
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Liyun Song
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Yinying Wang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Yao Bian
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Yong Qiu
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650000, People’s Republic of China
- Engineering Research Center of Classic Formula Regulate Immunity in Chronic Disease Prevention and Treatment, Kunming, Yunnan, 650000, People’s Republic of China
| |
Collapse
|
87
|
Dai Y, Vgontzas AN, Chen L, Zheng D, Chen B, Wu J, Shao R, Li Y. A multi-omics study of the association between insomnia with objective short sleep duration phenotype and high blood pressure. Sleep 2025; 48:zsaf030. [PMID: 39888642 DOI: 10.1093/sleep/zsaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/26/2025] [Indexed: 02/01/2025] Open
Abstract
STUDY OBJECTIVES Insomnia with objective short sleep duration is associated with increased hypertension risk. We aimed to explore the mechanism underlying the association between objective short sleep duration and hypertension in patients with chronic insomnia disorder (CID) by multi-omics. METHODS CID was defined according to International Classification of Sleep Disorders-3, and objective short sleep duration was based on the median value of total sleep time of the overall subjects during an overnight polysomnography. We used the mean values of measured nighttime and morning systolic (SBP) and diastolic blood pressure (DBP) for analysis. Serum metabolomics and fecal 16S rDNA amplicon sequencing were used to explore characteristic metabolites and analyze gut microbiota distribution, respectively. RESULTS One hundred and three patients with CID and 70 normal sleepers were included. We found 52 objective short sleep duration insomnia phenotype (ISSD)-related serum metabolites. Among the 52 ISSD-related serum metabolites, indoxyl sulfate was positively correlated with BP after adjusting for confounding factors (SBP: β = 0.250, p = .028; DBP: β = 0.256, p = .030) in ISSD. In addition, the level of serum indoxyl sulfate was significantly correlated with the genera Prevotella 9 (r = .378, p = .027), CAG-56 (r = -.359, p = .037), Ruminiclostridium 9 (r = -.340, p = .049), and Ruminococcus 2 (r = -.356, p = .039) in ISSD. CONCLUSIONS Our study suggests that the ISSD phenotype is associated with significant changes in serum metabolic profile, including high levels of indoxyl sulfate. The latter molecule correlates both with BP and gut microbiota in patients with the ISSD phenotype, suggesting that indoxyl sulfate may be the molecular path resulting in increased hypertension risk in this phenotype.
Collapse
Affiliation(s)
- Yanyuan Dai
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Alexandros N Vgontzas
- Sleep Research and Treatment Center, Department of Psychiatry and Behavioral Health, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Le Chen
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Dandan Zheng
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Baixin Chen
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Jun Wu
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Ruifan Shao
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Yun Li
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| |
Collapse
|
88
|
Peng B, Dai J, Ji S, Yang Y, Zuo A, Xu S, Fang W, Li D, You Y, Jiang Z, Wu C, Kwan HY, Zhao X, Liu Y. Quercetin ameliorates hyperuricemic nephropathy through improving gut dysfunctions and decreasing gut bacteria-derived uremic toxins. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156801. [PMID: 40403599 DOI: 10.1016/j.phymed.2025.156801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/23/2025] [Accepted: 04/21/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Hyperuricemia (HUA) is closely associated with gut dysbiosis, yet the role of microbial metabolism in hyperuricemic nephropathy (HN) remains poorly understood. Quercetin has shown urate-lowering and nephroprotective effects, but its therapeutic mechanisms, particularly in modulating the gut microbiome and microbial metabolism, remain elusive. PURPOSE This study investigates the therapeutic effects of quercetin on HN and explores its role in regulating host-microbial co-metabolism. METHODS A spontaneous HUA rat model (Uox-/- rats) was used to evaluate the therapeutic effect of quercetin. Multi-omics analyses, including gut microbiome profiling, peripheral untargeted metabolome, and targeted quantification of gut bacteria-derived uremic toxins, were performed. An integrated network analysis was conducted to uncover potential host-microbe metabolic interactions. RESULTS Quercetin treatment significantly reduced serum uric acid, creatinine, and blood urea nitrogen, ameliorated renal inflammation, fibrosis and oxidative stress, and improved gut dysbiosis and intestinal barrier dysfunction. Notably, high-dose quercetin downregulated Blautia, a key gut bacterium associated with uremic toxin production, and suppressed microbial phenylalanine metabolism, leading to decreased levels of gut bacteria-derived nephrotoxic metabolites (e.g., 3-phenyllactic acid, hippuric acid, and N-acetyl-l-phenylalanine). These uremic toxins were positively correlated with markers of kidney injury and proinflammatory cytokines. Mechanistically, quercetin modulated microbial enzymatic pathways involved in phenylalanine metabolism, thereby disrupting the formation of nephrotoxic metabolites and alleviating renal damage. CONCLUSIONS This study provides multi-omics evidence that quercetin ameliorates HN by regulating gut dysfunctions and decreasing gut bacteria-derived uremic toxins through host-microbial co-metabolism. These findings highlight the therapeutic potential of microbiota-targeted interventions in HUA-associated kidney diseases.
Collapse
Affiliation(s)
- Baizhao Peng
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaojiao Dai
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shuai Ji
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ying Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Anna Zuo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shanhua Xu
- Department of Pediatrics, Tianhe Traditional Chinese Medicine Hospital, Guangzhou, Guangdong 510665, China
| | - Wen Fang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Dexian Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yanting You
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zihao Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chuanghai Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoshan Zhao
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yanyan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
89
|
Wang C, Jiang X, Han S, Zang H, Gao X. Loganin ameliorates left ventricular fibrosis and dysfunction induced by pressure overload via the Sirt1/AKT/TGF-β1 signaling pathway. J Nat Med 2025:10.1007/s11418-025-01911-9. [PMID: 40347371 DOI: 10.1007/s11418-025-01911-9] [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: 01/26/2025] [Accepted: 04/18/2025] [Indexed: 05/12/2025]
Abstract
Loganin (LG), a natural compound derived from Cornus officinalis Sieb. et Zucc., possesses diverse pharmacological properties, such as anti-inflammatory, anti-hypertrophic, and antioxidant effects. However, the role of LG in the pathogenesis of Heart Failure (HF) remains unclear. The current work aimed to explore the underlying mechanism of LG in pressure overload-induced HF, both in vivo and in vitro, using transverse aortic constriction (TAC) surgery or isoproterenol (ISO) administration. Following eight weeks of TAC surgery, histological assessments, including hematoxylin and eosin staining, wheat germ agglutinin staining, TUNEL assay, and Masson's trichrome staining, were conducted to evaluate the extent of cardiomyocyte remodeling. Additionally, RT-PCR and WB analyses were performed to detect the levels of various targets. Furthermore, H9C2 cardiomyocytes were treated with ISO to induce hypertrophy, and the effects of LG on cell viability, α-smooth muscle actin (α-SMA) expression, and molecular targets were investigated. Our findings revealed that LG treatment at 40 mg/kg/day significantly attenuated cardiac dysfunction, decreased left ventricular collagen deposition in both interstitial and perivascular spaces. Mechanistically, LG mitigated ISO-induced toxicity in H9C2 cardiomyocytes, decreasing cellular hypertrophy and α-SMA expression. Moreover, we observed a downregulation of Sirtuin 1 (Sirt1) at the molecular level, accompanied by reduced phosphorylation of Akt and transforming growth factor-β1 (TGF-β1). Notably, the administration of the Sirt1 inhibitor, EX527, effectively abolished the protective effects of LG. Therefore, the cardio-protective effects of LG were mediated through the activation of the Sirt1/Akt/TGF-β1 signaling pathway, leading to reduced fibrosis and improved cardiac function.
Collapse
Affiliation(s)
- Changbin Wang
- Department of Cardiology, The First Rongjun Youfu Hospital of Shandong Province, No. 23 Jiefang Road, Jinan, 25000, China
| | - Xiaoli Jiang
- Department of Cardiology, The First Rongjun Youfu Hospital of Shandong Province, No. 23 Jiefang Road, Jinan, 25000, China
| | - Shuhua Han
- Station for Fengming Community Health Service, People's Hospital of Zhoucun District, No. 999 Hengxing Road, Zibo, 255300, China
| | - Huimei Zang
- Center for Gene and Immunotherapy, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.44 Wenhua West Road, Jinan, 250033, China
- Multidisciplinary Innovation Center for Nephrology of the Second Hospital, Cheeloo College of Medicine, Shandong University, No.44 Wenhua West Road, Jinan, 250033, China
| | - Xiaoyuan Gao
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Jinan, 250033, China.
| |
Collapse
|
90
|
Hou R, Shui W, Xing X, Chen Y, Shen W, Song Q, Wang J. Relationship Between TGF-β1 and Left Ventricular Geometry and Function in Patients With Essential Hypertension. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025. [PMID: 40346927 DOI: 10.1002/jcu.24055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/12/2025]
Abstract
OBJECTIVE To explore the association of transforming growth factor β1 (TGF-β1) with left ventricular geometry (LVG) and left ventricular function (LVF) in cases having essential hypertension. METHODS This retrospective study of 213 cases of essential hypertension, according to echocardiogram measurements, were split into normal geometry (NG), concentric remodeling (CR), eccentric hypertrophy (EH), and concentric hypertrophy (CH) groups. General clinical data of each patient was analyzed and office blood pressure measurements were performed. Detection of blood biochemistry and serum TGF-β1 content was conducted. The association of TGF-β1 with LVG and LVF parameters was assessed. RESULTS In contrast to the NG and CR groups, the TGF-β1 concentration was higher in the EH and CH groups, and it was most pronounced in the CH group. The TGF-β1 was positively linked to E/e' (r = 0.506, p < 0.001), whereas left ventricular global longitudinal strain (GLS) (r = -0.447, p < 0.001) was negatively correlated. Moreover, serum TGF-β1 levels were independently linked to EH and CH. CONCLUSION TGF-β1 was associated with abnormal LVG and LVF in cases of essential hypertension, indicating that it may induce LVG and LVF abnormalities.
Collapse
Affiliation(s)
- Ran Hou
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wen Shui
- Department of Cardiopulmonary Function Examination, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xueqing Xing
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaodong Chen
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenqian Shen
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qingfei Song
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jian Wang
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
91
|
Wang X, Wang J, Zheng B, Tian R, Huang L, Mao W, Feng Y, Liu B, Xu P. Sanqi oral solution alleviates podocyte apoptosis in experimental membranous nephropathy by mediating EMT through the ERK/CK2-α/β-catenin pathway. Front Pharmacol 2025; 16:1503961. [PMID: 40417210 PMCID: PMC12098599 DOI: 10.3389/fphar.2025.1503961] [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: 09/30/2024] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Sanqi oral solution (SQ) is a Chinese medicine that has been used well to treat idiopathic membranous nephropathy (IMN). It has been demonstrated to mitigate IMN proteinuria by inhibiting podocyte apoptosis. however, the precise mechanism has not been fully elucidated. Methods A passive Heymann nephropathy (PHN) rat model was used to mimic the in vivo disease characteristics of IMN. The PHN rats were intragastrically administered SQ (12.6/6.3 mL/kg) or tacrolimus (0.315 mg/kg) for 21 days. SQ was applied to ADR-induced podocytes in vitro. The effects of SQ on IMN and its underlying mechanisms were determined by measuring biochemical indices, pathomorphological characteristics, membrane attack complex (MAC), cell morphology, and protein levels. Results The SQ ingredients found in rat serum underscored their successful absorption in rats. In PHN rats, SQ induced a significant reduction in proteinuria, MAC, C5b-9, and glomerular basement membrane thickness, along with a drop in apoptotic podocytes. Similarly, SQ exerted a protective effect against ADR-induced podocyte injury by inhibiting apoptosis. Furthermore, inhibition of the ERK/CK2-α/β-catenin pathway-mediated epithelial-to-mesenchymal transition (EMT) was found to be involved in the anti-apoptotic effect of SQ in PHN rats and podocytes, marked by the reduction in vimentin and α-SMA and the induction of Synaptopodin and Podocin protein levels. Conclusion Inhibition of EMT via the ERK/CK2-α/β-catenin pathway may be the main mechanism by which SQ suppresses podocyte apoptosis in IMN.
Collapse
Affiliation(s)
- Xiaowan Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Juanjuan Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Bidan Zheng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruimin Tian
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Lihua Huang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Wei Mao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, China
| | - Yi Feng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Bo Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Peng Xu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, China
| |
Collapse
|
92
|
Jiao Q, Xu X, Xu L, Wang Y, Pang S, Hao J, Liu X, Zhao Y, Qi W, Qin L, Huang T, Li J, Wang T. Knockdown of eIF3a alleviates pulmonary arterial hypertension by inhibiting endothelial-to-mesenchymal transition via TGFβ1/SMAD pathway. J Transl Med 2025; 23:524. [PMID: 40346622 PMCID: PMC12065328 DOI: 10.1186/s12967-025-06505-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/13/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVE Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by vascular remodeling and involves Endothelial-to-Mesenchymal transition (EndMT) in pulmonary artery endothelial cells (PAECs). EndMT is a complex cell differentiation process, mainly showing the detachment of endothelial cell migration and reducing endothelial cell characteristics to varying degrees, acquiring mesenchymal cell characteristics. In addition, numerous studies have reported that eIF3a over expression plays an important role in the occurrence and development of fibrotic diseases, cancer, and degenerative lesions, however, the mechanisms of eIF3a affecting the dysfunction of pulmonary arterial endothelial cells remains largely unknown. Therefore, we aimed to demonstrate the underlying mechanisms of eIF3a-knockdown inhibiting EndMT by regulating TGFβ1/SMAD signal pathway in PAH. METHODS In this study, we screened the potential target genes associated with idiopathic pulmonary arterial hypertension (IPAH) by WGCNA to provide a reference for the diagnosis and treatment of PAH. By constructing WGCNA, which indicated that the blue module (module-trait associations between modules and clinical feature information were calculated to selected the optimum module) is most closely associated with IPAH, we further screened out 10 up-regulated candidate biomarker genes. Male SD rats were randomly assigned to four groups: Control, Monocrotaline (MCT), AAV1-shRNA-NC group and AAV1-shRNA-eIF3a group. The eIF3a-knockdown rat model was constructed by adeno-associated virus type-1 (AAV1) infection, PAH was evaluated according to hemodynamic alteration, right heart hypertrophy and histopathological changes in the lung tissue. Hematoxylin eosin (H&E) staining was used to assess the morphological changes of pulmonary arteries in rats of each treatment group. Co-localization of eIF3a with alpha-small muscle action (α-SMA) and co-localization of eIF3a with endothelial marker (CD31) were detected by double-label immunofluorescence. Immunohistochemistry (IHC) and Western blot (WB) experiments were performed to assess the expression of eIF3a, EndMT and TGFβ1/SMAD signal related proteins. In vitro, primary rat pulmonary artery endothelial cells (PAECs) were transfected with si-eIF3a to investigate the effects of eIF3a-knockdown on hypoxia-induced EndMT in PAECs and further elucidate its underlying molecular mechanisms. RESULTS By WGCNA analysis, we screened the up-regulated hub genes of TMF1, GOLGB1, ARMC8, PRPF40 A, EIF3 A, ROCK2, EIF5B, CCP110, and KRR1 associated with PAH, and in order to verify the potential role of eIF3a in the development of pulmonary arterial hypertension, MCT-induced PAH rat model was constructed successfully. The expression of eIF3a was increased in MCT-treated lungs. Knockdown of eIF3a significantly inhibited the pulmonary arterial hypertension and vascular remodeling in MCT-induced PAH rat model, ameliorated MCT-induced increases of right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (RVH) in rats. Double-labeled immunofluorescence showed eIF3a was mostly co-localized with CD31, this result indicated that the development of MCT-induced PAH was related to the regulation of PAECs function (most likely associated with the change of EndMT in endothelial cells). WB showed that the expressions of EndMT related proteins were significantly increased by regulating TGFβ1/SMAD signaling pathway in MCT-induced PAH rat lung tissues, however, knockdown of eIF3a markedly attenuated these changes. In addition, we observed the same results in rat PAECs with chronic hypoxia exposure. These results indicate that eIF3a-knockdown inhibited EndMT by regulating TGFβ1/SMAD signaling pathway in PAECs, thereby improving the development of MCT-induced PAH. CONCLUSIONS Knockdown of eIF3a inhibited EndMT in PAECs regulating TGFβ1/SMAD signaling pathway, significantly alleviated the changes of RVSP, RVH and vascular remodeling in MCT-induced PAH rats, eIF3a may be a promising and novel therapeutic target for the treatment of PAH.
Collapse
Affiliation(s)
- Qiuhong Jiao
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Xiufeng Xu
- Department of Geriatrics, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Longwu Xu
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Yuying Wang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Shulan Pang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Jie Hao
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaohong Liu
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Yudan Zhao
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Wanpeng Qi
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Limin Qin
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Tao Huang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China.
| |
Collapse
|
93
|
Li Q, Chen H, Gao H, Wei X, Bian D, Zheng L, Wei H, Wang W, Wang Y, Deng W. Role of CD33 basophils in mediating the effect of lipidome on chronic kidney disease: A 2-sample, 2-variable, bidirectional Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e42332. [PMID: 40355216 PMCID: PMC12073931 DOI: 10.1097/md.0000000000042332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/16/2025] [Indexed: 05/14/2025] Open
Abstract
This study aimed to investigate the causal relationship between lipidomes and chronic kidney disease (CKD) and identify and quantify the role of immune cells as a potential mediator. Using summary-level data from a genome-wide association study, a 2-sample Mendelian randomization (MR) analysis of genetically predicted lipidomes (7174 cases) and CKD (406,745 cases) was performed. Furthermore, we used 2-step MR to quantitate the proportion of the effect of immune cells traits-mediated lipidomes on CKD. The MR analysis revealed a causal relationship between lipidomes and CKD, with different lipidomes either increasing or decreasing the risk of CKD. Immune cells may serve as intermediaries in the pathway from lipidomes to CKD. Our study indicates that CD33 on basophils accounts for 3.23% of the reduced risk associated with triacylglycerol (53:3) levels in CKD. In conclusion, our study has identified a causal relationship between lipidomes and CKD, as well as the mediating role of CD33 on basophils. However, other risk factors like potential mediators require further investigation. In clinical practice, particular attention should be paid to lipidomic changes, especially triacylglycerol, in patients with CKD.
Collapse
Affiliation(s)
- Qi Li
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Haoyu Chen
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Hui Gao
- Department of Nephropathy, Hebei Province Hospital of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Xiaona Wei
- Department of Nephropathy, Hebei Province Hospital of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Dong Bian
- Department of Nephropathy, Hebei Province Hospital of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Linlin Zheng
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Hongyu Wei
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Wanqing Wang
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Yashi Wang
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| | - Wanying Deng
- School of Graduate, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, China
| |
Collapse
|
94
|
Cai N, Zhu SY, Huang JJ, Chen YX, Huang C, Qin XH. Rituximab, tacrolimus, cyclophosphamide and cyclosporin in primary membranous nephropathy with nephrotic syndrome: comparison of safety profiles, effect on remission rate, 24-h urinary total protein, serum albumin, and serum creatinine levels using network meta-analysis. Int Urol Nephrol 2025:10.1007/s11255-025-04549-4. [PMID: 40338506 DOI: 10.1007/s11255-025-04549-4] [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: 01/17/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE To compare the efficacy and safety of four immunosuppressive therapies, either alone or in combination, for primary membranous nephropathy through a network meta-analysis. METHODS A literature search was conducted for randomized controlled trials (RCTs) of Cyclophosphamide (CTX), Cyclosporin (CsA), Tacrolimus (TAC), and Rituximab (RIT) in the treatment of primary membranous nephropathy. Two researchers independently screened articles, extracted data, and evaluated the quality. Outcome indicators included dichotomous variables and continuous variables, which were represented by risk ratios (RR) and mean differences (MD), respectively. Then, various interventions were ranked according to the surface under the cumulative ranking curve (SUCRA). RESULTS A total of 21 randomized controlled trials (RCTs) were included, encompassing 1396 patients. In terms of the overall response rate (ORR), RIT+TAC was superior to CsA (RR = 0.15, 95% CI: 0.04, 0.54), CTX (RR = 0.09, 95% CI: 0.03, 0.31), and RIT (RR = 7.06, 95% CI: 2.29, 21.80). The SUCRA value of RIT+TAC was the highest, reaching 93.5%. Regarding the total 24-h urinary protein (24UTP), RIT+TAC was better than RIT (MD = 17.05, 95% CI: 6.49, 44.79), RIT+CTX (MD = 6.99, 95% CI: 2.55, 19.17), TAC (MD = 0.12, 95% CI: 0.07, 0.18), CsA (MD = 0.06, 95% CI: 0.00, 0.86), and CTX (MD = 0.05, 95% CI: 0.03, 0.10). The SUCRA value of RIT+TAC was the highest, at 99.4%. For serum albumin, RIT+CTX was superior to CTX (MD = 0.00, 95% CI: 0.00, 0.29), and the SUCRA value of RIT+CTX was the highest, at 76.7%. For serum creatinine (Scr), RIT+TAC was better than TAC (MD = 0.00, 95% CI: 0.00, 0.13), and CsA was better than TAC (MD = 7.86e+07, 95% CI: 3.65e+06, 1.69e+09). The SUCRA value of RIT+TAC was the highest, at 79.9%. In terms of the incidence of adverse reactions, CTX had a higher rate than RIT+CTX (RR = 11.12, 95% CI: 1.34, 92.15). The SUCRA value of RIT+CTX was the lowest, at 5.3%. CONCLUSION In terms of improving ORR, reducing 24UTP and lowering Scr, the RIT+TAC regimen may be the most optimal. Conversely, RIT+CTX demonstrated the best efficacy in improving ALB and also exhibited relatively better safety profile.
Collapse
Affiliation(s)
- Ni Cai
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, Jiangxi, China
| | - Shu-Ying Zhu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, Jiangxi, China
| | - Jin-Jing Huang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, Jiangxi, China
| | - Yan-Xia Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, Jiangxi, China
| | - Chong Huang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, Jiangxi, China
| | - Xiao-Hua Qin
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
95
|
Jiang M, Bu W, Wang X, Ruan J, Shi W, Yu S, Huang L, Xue P, Tang J, Zhao X, Su L, Cheng D. Pulmonary fibrosis: from mechanisms to therapies. J Transl Med 2025; 23:515. [PMID: 40340941 PMCID: PMC12063347 DOI: 10.1186/s12967-025-06514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive interstitial lung disease characterized by excessive deposition of extracellular matrix (ECM) and abnormal fibroblast proliferation, which is mainly caused by air pollution, smoking, aging, occupational exposure, environmental pollutants exposure, and microbial infections. Although antifibrotic agents such as pirfenidone and nintedanib, approved by the United States (US) Food and Drug Administration (FDA), can slow the decline in lung function and disease progression, their side effects and delivery inefficiency limit the overall prognosis of PF. Therefore, there is an urgent need to develop effective therapeutic targets and delivery approaches for PF in clinical settings. This review provides an overview of the pathogenic mechanisms, therapeutic drug targeting signaling pathways, and promising drug delivery strategies for treating PF.
Collapse
Affiliation(s)
- Mengna Jiang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Wenxia Bu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xuehai Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Jialing Ruan
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Weijian Shi
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Siqi Yu
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Lizhen Huang
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Peng Xue
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| | - Liling Su
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China.
| | - Demin Cheng
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| |
Collapse
|
96
|
Wang L, Qiu T, Xu F, Zhang L, Zhang C, Ye W. Fabricate heparin-mimic thin gel layers for vascular cell selective regulation using 5-hydroxydopamine cross-linked chitosan and sulfonated polymers. Int J Biol Macromol 2025; 311:144027. [PMID: 40345300 DOI: 10.1016/j.ijbiomac.2025.144027] [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: 02/17/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
In this work, 5-hydroxydopamine was employed as a crosslinking agent to bind chitosan and sulfonated polymers to fabricate thin gel layers (TGLs) featuring heparin-mimic structures. By controlling the distribution of exposed chemical moieties (phenolic hydroxyl, amino, and sulfonic groups), the growth of the endothelial cell (EC) and smooth muscle cell (SMC) on the TGLs surfaces could be modulated. Such modulation effectively maintained the quantity and proportion of the two cell types within a reasonable range, thereby offering a potential avenue for promoting re-endothelialization. The prepared TGLs showed improved hydrophilicity as well as hemocompatibility. For cytocompatibility test, TGLs led to a notable promotion of the growth of human umbilical vein endothelial cells (HUVECs) and exerted substantial inhibitory effects on the proliferation of human umbilical artery smooth muscle cells (HUASMCs). The ratio of HUVECs to HUASMCs rose from 0.184 to 1.97. The enhanced hemocompatibility was attributed to the incorporation of exposed functional groups. Regarding the highly selective effects, these were ascribed to the synergistic influence of high sulfonation degree and the presence of amino groups and phenolic hydroxyl groups. The current work illustrated a simple method for synthesizing a multifunctional biomimetic polymer material that offers the promise of broader biomedical research applications.
Collapse
Affiliation(s)
- Lingren Wang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China.
| | - Tingting Qiu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Fan Xu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Linna Zhang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Chao Zhang
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China
| | - Wei Ye
- Department of Chemical Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu 223003, China.
| |
Collapse
|
97
|
Huang H, Ru SJ, Chen JM, Liu W, Fang SH, Liu Q, Meng Q, Liu P, Zhou H. Quantitative Proteomic Study Reveals Amygdalin Alleviates Liver Fibrosis Through Inhibiting mTOR/PDCD4/JNK Pathway in Hepatic Stellate Cells. Drug Des Devel Ther 2025; 19:3735-3749. [PMID: 40356680 PMCID: PMC12067723 DOI: 10.2147/dddt.s500439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/05/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose Hepatic fibrosis is a major cause of morbidity and mortality for which there is currently limited therapy. Amygdalin, a cyanogenic glucoside derived from Semen Persicae, exerts significant anti-fibrotic effects in the liver. However, the molecular mechanism by which amygdalin inhibits the progression of liver fibrosis remains unclear. This study aimed to elucidate the potential mechanism of action of amygdalin against liver fibrosis. Methods Quantitative proteomic profiling of the mouse liver tissues from control, carbon tetrachloride (CCl4)-induced fibrosis, and amygdalin-treated groups was performed to explore the key effector proteins of amygdalin. Histology and immunohistochemistry as well as serum biochemical analysis were performed to evaluate amygdalin efficacy in mice. The key gene programmed cell death protein 4 (PDCD4) was overexpressed or knocked down in human hepatic stellate cells (HSCs). The mRNA and protein levels of related molecules were detected by RT-qPCR and Western blotting, respectively. Results Amygdalin could effectively ameliorated CCl4-induced liver fibrosis in mice. Bioinformatics analysis revealed that PDCD4 was downregulated in CCl4-induced liver fibrosis, but amygdalin treatment reversed these changes. An in vitro study showed that PDCD4 inhibited the activation of human hepatic stellate cell line LX-2 cells by regulating the JNK/c-Jun pathway and amygdalin inhibited the activation of LX-2 cells in a PDCD4-dependent manner. We further found that amygdalin inhibited the phosphorylation of PDCD4 at Ser67 by inhibiting the mTOR/S6K1 pathway to enhance PDCD4 expression. Conclusion Our data demonstrated a potential pharmaceutical mechanism by which amygdalin alleviates liver fibrosis by inhibiting the mTOR/PDCD4/JNK pathway in HSCs, suggesting that PDCD4 is a potential target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Hui Huang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People’s Republic of China
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Su-Jie Ru
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Jia-Mei Chen
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, People’s Republic of China
| | - Wei Liu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, People’s Republic of China
| | - Shan-Hua Fang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Qian Meng
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Ping Liu
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, People’s Republic of China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hu Zhou
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, People’s Republic of China
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
98
|
Xia LY, Yu NR, Huang SL, Qu H, Qin L, Zhao QS, Leng Y. Dehydrotrametenolic acid methyl ester, a triterpenoid of Poria cocos, alleviates non-alcoholic steatohepatitis by suppressing NLRP3 inflammasome activation via targeting Caspase-1 in mice. Acta Pharmacol Sin 2025:10.1038/s41401-025-01569-9. [PMID: 40329004 DOI: 10.1038/s41401-025-01569-9] [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: 01/14/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025]
Abstract
Non-alcoholic steatohepatitis (NASH) has emerged as a prevalent chronic liver disease with a huge unmet clinical need. A few studies have reported the beneficial effects of Poria cocos Wolf (P. cocos) extract on NASH mice, but the active components were still unknown. In this study we investigated the therapeutic effects of dehydrotrametenolic acid methyl ester (ZQS5029-1), a lanosterol-7,9(11)-diene triterpenes in P. cocos, in a high-fat diet plus CCl4 induced murine NASH model and a GAN diet induced ob/ob murine NASH model. The NASH mice were treated with ZQS5029-1 (75 mg·kg-1·d-1, i.g.) for 6 and 8 weeks, respectively. We showed that ZQS5029-1 treatment markedly relieved liver injury, inflammation and fibrosis in both the murine NASH models. We found that ZQS5029-1 treatment significantly suppressed hepatic NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation in both the NASH murine models, and blocked lipopolysaccharides (LPS)+adenosine 5'-triphosphate (ATP)/Nigericin-induced NLRP3 inflammasome activation in bone marrow-derived macrophages (BMDMs) and Kupffer cells in vitro. We demonstrated that ZQS5029-1 directly bound to the H236 residue of mouse Caspase-1, thereby inhibiting NLRP3 inflammasome activation. The effects of ZQS5029-1 on macrophage-hepatocyte/HSC crosstalk were analyzed using the supernatants from macrophages preconditioned with LPS + ATP introduced into hepatocytes and hepatic stellate cells (HSCs). We found that the conditioned medium from the BMDMs induced injury and death, as well as lipid accumulation in hepatocytes, and activation of HSCs; these effects were blocked by conditioned medium from BMDMs treated with ZQS5029-1. Moreover, the protective effects of ZQS5029-1 on hepatocytes and HSCs were eliminated by H236A-mutation of Caspase-1. We conclude that ZQS5029-1 is a promising lead compound for the treatment of NASH by inhibiting NLRP3 inflammasome activation through targeting Caspase-1 and regulating the macrophage-hepatocyte/HSC crosstalk.
Collapse
Affiliation(s)
- Ling-Yan Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nai-Rong Yu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Su-Ling Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hui Qu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Qin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qin-Shi Zhao
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Ying Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
99
|
Elnaggar JH, Lammons JW, Ardizzone CM, Aaron KJ, Jacobs C, Graves KJ, George SD, Luo M, Tamhane A, Łaniewski P, Quayle AJ, Herbst-Kralovetz MM, Cerca N, Muzny CA, Taylor CM. Predicting Bacterial Vaginosis Development using Artificial Neural Networks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.02.25326872. [PMID: 40385447 PMCID: PMC12083586 DOI: 10.1101/2025.05.02.25326872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Bacterial vaginosis (BV) is a dysbiosis of the vaginal microbiome, characterized by the depletion of protective Lactobacillus spp. and overgrowth of anaerobes. Artificial neural network (ANN) modeling of vaginal microbial communities offers an opportunity for early detection of incident BV (iBV). 16S rRNA gene sequencing and quantitative PCR was performed on longitudinal vaginal specimens collected from participants within 14 days of iBV or from healthy participants to calculate the inferred absolute abundance (IAA) of vaginal bacterial taxa. ANNs were trained using the IAA of vaginal taxa from 420 vaginal specimens to classify individual vaginal specimens as either pre-iBV (collected before iBV onset) or Healthy. Feature importance was assessed to understand how specific vaginal micro-organisms contributed to model predictions. ANN modeling accurately classified >97% of specimens as either pre-iBV or Healthy (sensitivity >96%, specificity >98%) using IAA of 20 vaginal taxa. Model prediction accuracy was maintained when training models using only a few key vaginal taxa. Models trained using only the top five most important features achieved an accuracy of >97%, sensitivity >92%, and specificity >99%. Model predictive accuracy was further improved by training models on specimens from white and black participants separately; using only three feature models achieved an accuracy >96%, sensitivity >91%, and specificity >91%. Feature analysis found that Lactobacillus species L. gasseri and L. jensenii differed in how they contributed to model predictions in models trained with data stratified by race. A total of 420 vaginal specimens were analyzed, providing a robust dataset for model training and validation.
Collapse
Affiliation(s)
- Jacob H. Elnaggar
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
| | - John W. Lammons
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
| | - Caleb M. Ardizzone
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
- Department of Microbiology, Indiana University School of Medicine; Indianapolis, IN, USA
| | - Kristal J. Aaron
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham; Birmingham, Alabama, USA
| | - Clayton Jacobs
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
| | - Keonte J. Graves
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham; Birmingham, Alabama, USA
| | - Sheridan D. George
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham; Birmingham, Alabama, USA
| | - Meng Luo
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
| | - Ashutosh Tamhane
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Alison J. Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
| | - Melissa M. Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Nuno Cerca
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira, Minho University, Braga, Portugal
| | - Christina A. Muzny
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham; Birmingham, Alabama, USA
| | - Christopher M. Taylor
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center; New Orleans, Louisiana, USA
| |
Collapse
|
100
|
Yao S, Marron MM, Tian Q, Watts EL, Clish CB, Shah RV, Murthy VL, Newman AB. Metabolomic Pathways of Inflammation and Mitochondrial Dysfunction Are Related to Worsening Healthy Aging Index and Mortality. J Gerontol A Biol Sci Med Sci 2025; 80:glaf057. [PMID: 40152499 DOI: 10.1093/gerona/glaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Metabolic-inflammatory states are central to multiorgan mechanisms of aging, but precise functional biomarkers of physiological aging remain less clear. METHODS In the Health, Aging, and Body Composition study, we defined metabolomic profiles of the Healthy Aging Index (HAI), a composite of cardiovascular, lung, cognitive, metabolic, and renal function (0-10, with higher scores indicating poorer health) in a split set design from 2015 older participants (mean age 73.6 years; 50% women; 35% Black). We used standard regression to identify metabolomic correlates of Year 1 and Year 10 HAI, change in HAI over time, and mortality. A metabolite score of HAI was developed using LASSO regression. RESULTS We identified 42 metabolites consistently associated with Year 1 and Year 10 HAI, as well as change in HAI: 13 lipids, 4 amino acids, and 4 metabolites of other classes were associated with worse and worsening HAI while 20 lipids and 1 amino acid was associated with better and improving HAI. Most of these associations were no longer significant after additionally adjusting for inflammation biomarkers. A higher metabolite score of Year 1 HAI was associated with greater HAI deterioration over time (hold-out "test" set beta 0.40 [0.15-0.65]) and higher mortality (hold-out "test" set hazard ratio: 1.43 [1.23-1.67]). CONCLUSIONS A multiorgan healthy aging phenotype was linked to lipid metabolites, suggesting potential pathways related to mitochondrial function, oxidative stress, and inflammation. Metabolomics of HAI at older age were related to worsening health and mortality, suggesting potential links between metabolism and accelerated physiological aging.
Collapse
Affiliation(s)
- Shanshan Yao
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Qu Tian
- National Institute of Aging, Baltimore, Maryland, USA
| | - Eleanor L Watts
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Ravi V Shah
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Anne B Newman
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|