51
|
Chen C, Li J, Liu F, Mu J, Gu X, Wei L, Dong J, Li M, Liu C, Ren Z, Tang W, Sun J, Chen X, Wang L, Yang S, Shen C. Construction and validation of a multi-mRNA panel and genetic scores for Krüppel-like factors for acute ischemic stroke. Int J Biol Macromol 2025:145222. [PMID: 40516726 DOI: 10.1016/j.ijbiomac.2025.145222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 06/02/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Cellular and animal experiments suggest that Krϋppel-like factors (KLFs) may be involved in the pathological mechanisms of acute ischemic stroke (AIS), but epidemiological research on KLFs in AIS is still limited. Therefore, we conducted two AIS case-control studies, and three prospective cohort studies to elucidate the contribution of 14 KLFs' distinct leukocytes mRNA expression (360 AIS cases vs. 363 controls) and 21 genetic tagSNPs (3651 AIS cases vs. 3900 controls) to the etiology and prognosis of AIS. We found KLFs mRNA score constructed by differentially expressed mRNAs significantly enhances the discriminative value for AIS compared to the traditional risk factors [the area under the curve (AUC) increased from 0.724 to 0.871]. Additionally, the KLFs polygenetic risk score could effectively stratify the susceptibility to AIS incidence (Ptrend < 0.001) and the highest PRS group has an increased 42.4 % risk of AIS onset in the cohort study. Furthermore, KLF12 mRNA negatively related to modified Rankin Scale (mRS) both at admission and discharge and KLF12 variants contributed to an elevated risk of IS death after AIS. In conclusion, this study provides new insights into the novel contribution of KLFs mRNA and genetic variants to the incidence risk and prognosis of AIS.
Collapse
Affiliation(s)
- Changying Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Li
- Department of Neurology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Fangyuan Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jialing Mu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xincheng Gu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lai Wei
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiayi Dong
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mengxia Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunlan Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhanyun Ren
- Department of Neurology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Wuzhuang Tang
- Department of Neurology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Junxiang Sun
- Department of Cardiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Xuemei Chen
- Department of Neurology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Wang
- Department of Neurology, Jurong Hospital Affiliated to Jiangsu University, Jurong People's Hospital, Jurong, Jiangsu, China
| | - Song Yang
- Department of Cardiology, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
52
|
Duan J, Pei F, Miao J, Liu S, Tan L, Lu M, Liu Y, Zhang C. Swietenine improved the progression of diabetic nephropathy through inhibiting ferroptosis via activating Akt/GSK-3β/Nrf2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119981. [PMID: 40378934 DOI: 10.1016/j.jep.2025.119981] [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: 12/30/2024] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Swietenia macrophylla King is a traditional medicinal plant extensively utilized in Asia and its pharmacological properties primarily involve antidiabetic, anti-inflammatory, antioxidant, antibacterial, and antitumor effects. Swietenine (Swi), the major bioactive compound presents in the fruits of S. macrophylla, has demonstrated beneficial therapeutic effects on diabetic nephropathy (DN). However, the underlying mechanism through which Swi influences DN remains unclear. AIM OF THE STUDY The current research aims to investigate the effects of Swi on DN and explore its underlying mechanisms associated with ferroptosis, both in vivo and in vitro. METHODS A model of streptozotocin/high-fat diet (STZ/HFD)-induced Sprague-Dawley (SD) rats was employed to assess the effect of Swi on improving DN and resisting ferroptosis in vivo. Additionally, mouse podocyte cells (MPC-5 cells) were induced by high glucose (HG) and cultured to explore the potential mechanisms of Swi in treating DN in vitro. To further validate the protective effects of Swi, pathway-specific inhibitors were administered to HG-induced MPC-5 cells to confirm the involvement of the Akt/GSK-3β/Nrf2 signaling pathway in the inhibition of ferroptosis. A combination of proteomics, immunohistochemical staining, western blotting, and cell culture techniques was utilized to explore the pharmacological mechanisms of Swi. Furthermore, network pharmacology and molecular docking analyses were conducted to predict the targets of Swi in relation to DN, which were subsequently validated through Western blotting analysis. RESULTS Administration of Swi significantly enhanced renal function and ameliorated pathological alterations in DN rats, as well as improved oxidative stress and inhibited ferroptosis. In vitro studies revealed that Swi dramatically improved the cell viability and mitigated oxidative stress, and inhibited ferroptosis via activating the Akt/GSK-3β/Nrf2 signaling pathway in HG-induced MPC-5 cells. CONCLUSION This study demonstrates that Swi improves DN by inhibiting ferroptosis via activating Akt/GSK-3β/Nrf2 signaling pathway for the first time, thereby providing a scientific basis that Swi is expected to be a promising candidate drug for the treatment of DN.
Collapse
Affiliation(s)
- Jingyu Duan
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Feilong Pei
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Jiale Miao
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Shuang Liu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Lin Tan
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Mengyuan Lu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Yaowu Liu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Chunping Zhang
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| |
Collapse
|
53
|
Ran L, Wei C, Wang X, Zhao Y, Li P, Huang D, Gu W, Wu X, Liang Z, Wang X, Gong L. SPHK2 inhibition alleviates chronic intermittent hypoxia-induced inflammation in adipose tissue by decreasing endoplasmic reticulum stress. Eur J Pharmacol 2025:177841. [PMID: 40513935 DOI: 10.1016/j.ejphar.2025.177841] [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: 03/18/2025] [Revised: 06/06/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Obstructive Sleep Apnea (OSA), characterized by chronic intermittent hypoxia (CIH), is a prevalent disorder that significantly elevates the risk of cardiovascular and metabolic complications. Lipid metabolism disturbances, a common comorbidity in OSA, are closely associated with CIH-induced adipose tissue inflammation, yet the underlying mechanisms remain poorly defined. In this study, we established a CIH mouse model to investigate the roles of Sphingosine Kinase 2 (SPHK2) and endoplasmic reticulum stress (ERS) in CIH-induced lipid metabolic disturbances. Mice were exposed to normal air (NA) or CIH for 4 or 12 weeks. Serum triglycerides (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) were measured to assess systemic lipid metabolism. CIH group significantly elevated SPHK2 expression and activated the PERK-ATF4-CHOP signaling pathway in epididymal (eWAT) and subcutaneous white adipose tissue (scWAT), leading to increased NLRP3 inflammasome accumulation, oxidative stress, and adipocyte apoptosis. Administration of the SPHK2 inhibitor ABC294640, opaganib, attenuated these effects, reducing ERS activation and restoring lipid homeostasis. Further, treatment with the ERS inhibitor 4-BPA suppressed oxidative stress and inflammatory cytokines, alleviating CIH-induced adipose tissue inflammation. In contrast, ERS activation by thapsigargin reversed the protective effects of SPHK2 inhibition, exacerbating metabolic and inflammatory dysregulation. In summary, our findings highlight the critical role of SPHK2 and ERS in CIH-induced adipose tissue inflammation and lipid metabolic disturbances. Pharmacological inhibition of SPHK2 represents a promising therapeutic approach for mitigating OSA-related lipid metabolic disturbances.
Collapse
Affiliation(s)
- Longyi Ran
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity
| | - Chang Wei
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity
| | - Xinyu Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity
| | - Yuean Zhao
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity
| | - Peijun Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity
| | - Dong Huang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity
| | - Wenyu Gu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301, Yanchang Rd., Shanghai, 200072, China
| | - Xu Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zongan Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity.
| | - Xinyuan Wang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Linjing Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Respiratory Health and Multimorbidity.
| |
Collapse
|
54
|
Shen S, Sugai-Munson A, Xiao X, Zhang Y, Kang J, Khetarpal SA, Jonas ER, Ambardekar AV, Bristow MR, Li H. Exercise Mitigates Diabetic Cardiomyopathy by Suppressing Ferroptosis via N-Acetyltransferase 10. Circ Res 2025. [PMID: 40492308 DOI: 10.1161/circresaha.125.326303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
Affiliation(s)
- Siman Shen
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
| | - Aya Sugai-Munson
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
| | - Xiao Xiao
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
| | - Yajing Zhang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., S.A.K., H.L.)
| | - Jiayi Kang
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
- Department of Physiology and Cellular Biophysics, Center for Molecular Cardiology, Columbia University, New York, NY (J.K.)
| | - Sumeet A Khetarpal
- Cardiovascular Research Center, Massachusetts General Hospital, Boston. (S.A.K.)
- Harvard Medical School, Boston, MA (Y.Z., S.A.K., H.L.)
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA (S.A.K.)
| | - Eric R Jonas
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Amrut V Ambardekar
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Michael R Bristow
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (E.R.J., A.V.A., M.R.B.)
| | - Haobo Li
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston. (S.S., A.S.-M., X.X., Y.Z., J.K., H.L.)
- Harvard Medical School, Boston, MA (Y.Z., S.A.K., H.L.)
| |
Collapse
|
55
|
Rani N, Arya DS. Modulation of PPAR-γ/Nrf2 and AGE/RAGE signaling contributes to the chrysin cardioprotection against myocardial damage following ischemia/reperfusion in diabetic rats. J Pharm Pharmacol 2025; 77:794-804. [PMID: 39673242 DOI: 10.1093/jpp/rgae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/15/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVE Advanced glycation end products/receptor for AGEs (AGE/RAGE) signaling has a well-established role in the etiology of diabetic-related cardiovascular disorders. The purpose of the study was to elucidate the role of chrysin, a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist, against ischemia/reperfusion (IR) injury in diabetic rats and its functional interaction with the AGE/RAGE signaling pathway. METHODS A single intraperitoneal injection of streptozotocin (STZ, 70 mg/kg) was administered to rats for induction of diabetes. Rats having blood glucose levels more than 300 mg/dl following a 72 hr STZ injection were classified as diabetic. PPAR-γ antagonist GW9662 (1 mg/kg, i.p.), chrysin (60 mg/kg, p.o.), or both were administered to diabetic rats for 4 weeks. On the 29th day, rats were given ischemia for 45 min and then reperfusion for 1 hr to induce myocardial infarction (MI). KEY FINDINGS Pretreatment with chrysin significantly improved hemodynamic status, ventricular functions, and cardiac injury markers in diabetic myocardium. Increased PPAR-γ/Nrf2 and decreased RAGE protein expressions were linked to this improvement. Chrysin pretreatment resulted in the upregulation of endogenous antioxidants and reduced TBARS levels. Moreover, chrysin significantly decreased inflammation and apoptosis in diabetic myocardium. CONCLUSION PPAR-γ/Nrf2 co-activation by chrysin ameliorated IR-induced MI in diabetic rats, possibly via modulating AGE/RAGE signaling.
Collapse
Affiliation(s)
- Neha Rani
- Department of Pharmacology, Kalpana Chawla Government Medical College, Karnal, Haryana, 132001, India
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
56
|
Lin Y, Yang S, Guo J. Antioxidant proteins can be potential targets in ameliorating ferroptosis in diabetic cardiomyopathy: a literature review. Diabetol Metab Syndr 2025; 17:199. [PMID: 40481517 PMCID: PMC12144738 DOI: 10.1186/s13098-025-01773-x] [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: 02/28/2025] [Accepted: 05/31/2025] [Indexed: 06/11/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the cardiovascular complications of diabetes mellitus, which is different from myocardial damage caused by coronary ischemia, hypertension, and valvular disease. DCM lacks distinct clinical manifestations in its early stages, and current therapeutic approaches primarily focus on symptomatic management. Emerging evidence indicates that even with optimized glycemic regulation, the pathophysiological progression of DCM remains unmitigated. Exploring the pathogenic mechanism of DCM is the focus and hotspot of current research. Ferroptosis, an iron-dependent form of regulatory cell death, is crucial in DCM myocardial damage. Dysfunctional antioxidant defense system, increased oxidative stress, and elevated reactive oxygen species are the key mechanisms of ferroptosis in DCM. Thus, this review innovatively takes antioxidant proteins as the entry point, and for the first time systematically summarizes the molecular mechanism of antioxidant proteins to improve DCM by regulating the ferroptosis pathway, and summarizes the therapeutic strategy of medications to enhance ferroptosis in DCM by targeting the expression of antioxidant proteins, to explore the potential targets to improve ferroptosis in DCM, to provide a new perspective for the study of delaying the progression of DCM.
Collapse
Affiliation(s)
- Yuting Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou, China
| | - Shu Yang
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jinjian Guo
- Department of Cardiology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou, China.
| |
Collapse
|
57
|
Li H, Li G, Gao Y, Ma Y, Yu Z, Zhang A, Yang G, Hou Z, Zhang Y, Yu Y, Zhang Z. Oligo-peptide I-C-F-6 mitigates polymicrobial sepsis-induced cardiac dysfunction in mice. Eur J Pharmacol 2025; 996:177545. [PMID: 40139420 DOI: 10.1016/j.ejphar.2025.177545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cardiomyopathy accounts for worse clinical outcome and higher mortality rate during sepsis globally. Here we assessed whether post-operative administration of I-C-F-6, a small molecule oligo-peptide (Gly-Ala-Gly-Pro-His-Gly-Gly) derived from Carapax trionycis, protected against septic cardiomyopathy in mice. Male adult mice were exposed to cecal ligation and puncture (CLP) and I-C-F-6 was administered intravenously (0.4 mg/kg or 4.0 mg/kg) 30 min following surgery. Administration of I-C-F-6 extended survival period and decreased sepsis severity score in septic mice. Furthermore, administration of I-C-F-6 mitigated cardiac atrophy and preserved cardiac function in septic mice. Mechanistically, I-C-F-6 inhibited inflammation and promoted M2 polarization in myocardium of septic mice. In addition, I-C-F-6 activated nuclear factor erythroid 2-related factor 2 (Nrf2)/haem oxygenase-1 (HO-1)/glutathione peroxidase 4 (GPX4) pathway, mitigated oxidative damage and inhibited ferroptosis in myocardium of septic mice. In conclusion, post-operative administration of I-C-F-6 in mice exposed to CLP improved survival and mitigated myocardial impairment. Our work established a clear therapeutic potential of I-C-F-6 for sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Hongxiao Li
- Department of Cardiology, Shanghai East Hospital, Tongji University, Jimo Road 150, Shanghai, 200120, China
| | - Guang Li
- Department of Emergency Trauma Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yuan Gao
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Yulin Ma
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Zixuan Yu
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Anna Zhang
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Guoling Yang
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Zhiqi Hou
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China.
| | - Zhigang Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University, Jimo Road 150, Shanghai, 200120, China.
| |
Collapse
|
58
|
Kowalski S, Wityk P, Raczak-Gutknecht J, Olszewska A, Żmijewski M, Kocić I. The imidazoline I 2 receptor agonist 2-BFI enhances cytotoxic activity of hydroxychloroquine by modulating oxidative stress, energy-related metabolism and autophagic influx in human colorectal adenocarcinoma cell lines. Eur J Pharmacol 2025; 996:177590. [PMID: 40185322 DOI: 10.1016/j.ejphar.2025.177590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Recently, interest in imidazoline receptors (IRs) has been increasing. Over the years, a growing number of studies have highlighted the therapeutic potential of ligands targeting these receptors, however, the potential role of imidazoline I2 receptor agonists in cancer treatment has not been thoroughly investigated. Colorectal cancer (CRC) is among the most prevalent and lethal forms of cancer worldwide. The complexity of CRC necessitates individualized approaches. One promising area of research within CRC therapy is the regulation of autophagy. Recent studies have explored the relationship between autophagy and cancer progression, revealing that autophagy modulation could be a potential strategy for CRC treatment. However, the mechanisms underlying autophagy regulation remain poorly understood. This study aimed to evaluate the effect of the imidazoline I2 receptor agonist, namely 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI), on colorectal cancer cells, HT-29 and HCT-116 cell lines, particularly its impact when co-incubated with the autophagy inhibitor, hydroxychloroquine (HCQ). The results showed that 2-BFI synergistically increased the cytotoxic effect of HCQ by inducing oxidative stress and apoptosis. Furthermore, our investigation indicated impairment autophagic influx in colon cancer cells treated by 2-BFI. The comprehensive metabolomic analysis revealed significant alterations in key metabolic pathways including MAO activity, oxidative stress responses, energy-related metabolites and amino acids metabolism. Altogether, these findings demonstrate potential a new therapeutic strategy based on autophagy regulation and the selective induction of oxidative stress in colorectal cancer cells. Moreover, this study provides a foundation for further investigation into the therapeutic potential of imidazoline receptor agonists in cancer therapy.
Collapse
Affiliation(s)
- Szymon Kowalski
- Department of Pharmacology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - Paweł Wityk
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland; Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, Gdansk, Poland; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, Gdansk, Poland
| | - Anna Olszewska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Żmijewski
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ivan Kocić
- Department of Pharmacology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
59
|
Pimenta RML, Skon-Hegg C, Rose-Hellekant T, Holy J. Mechanoresponsive patterns of KLF2, 4, 5, and 6 expression differ among subclones from a single mammary tumor. Acta Histochem 2025; 127:152238. [PMID: 39983249 DOI: 10.1016/j.acthis.2025.152238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
A number of Krüppel-like transcription factor (KLF) family members display mechanoresponsive behaviors, and function as mechanosensitive transcription factors. There are many normal and pathological conditions where their roles in mechanotransduction and mechanoadaptation are not well understood, however. In this study, two basic questions regarding KLF mechanoresponsiveness were addressed: 1) are KLF 2, 4, 5, and 6 expressed at different levels among subclones of tumor cells adapted to specific microenvironmental conditions; and 2) is the expression of these KLFs responsive to rapid changes in the physical environment? To address these questions, the heterogeneous and differentially metastatic murine mammary tumor subclones 4T1, 4T07, and 67NR were subjected to physical changes in their culture conditions, and KLF responses assessed. The results show that the expression of different KLFs exhibit distinct responses to reductions in cell tension, as well as cell detachment from 2D and 3D environments. KLF2 and 4 expression is rapidly and temporarily induced upon release of cells from a stiff 2D substrate into liquid suspension culture in all three subclones, and similar responses are observed in two of the subclones upon the release of tension in 3D collagen gel cultures. By contrast, expression patterns of KLF5 and 6 were generally less affected by physical changes in most, but not all, of the cell lines examined. These results support the concept that KLFs differentially participate in transducing physical differences among intratumoral neighborhoods into distinct responses among heterogeneous subclones, thereby contributing to tumor cell behavioral complexity.
Collapse
Affiliation(s)
| | - Cara Skon-Hegg
- Whiteside Institute for Clinical Research, St. Luke's Hospital, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Teresa Rose-Hellekant
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Jon Holy
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA.
| |
Collapse
|
60
|
Mahmood NMS, Mahmud AMR, Maulood IM. Vascular actions of Ang 1-7 and Ang 1-8 through EDRFs and EDHFs in non-diabetes and diabetes mellitus. Nitric Oxide 2025; 156:9-26. [PMID: 40032212 DOI: 10.1016/j.niox.2025.02.003] [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: 10/14/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in regulating vascular homeostasis, while angiotensin 1-8 (Ang 1-8) traditionally dominates as a vasoconstrictor factor. However, the discovery of angiotensin 1-7 (Ang 1-7) and Ang 1-8 has revealed counter-regulatory mechanisms mediated through endothelial-derived relaxing factors (EDRFs) and endothelial-derived hyperpolarizing factors (EDHFs). This review delves into the vascular actions of Ang 1-7 and Ang 1-8 in both non-diabetes mellitus (non-DM) and diabetes mellitus (DM) conditions, highlighting their effects on vascular endothelial cell (VECs) function as well. In a non-DM vasculature context, Ang 1-8 demonstrate dual effect including vasoconstriction and vasodilation, respectively. Additionally, Ang 1-7 induces vasodilation upon nitric oxide (NO) production as a prominent EDRFs in distinct mechanisms. Further research elucidating the precise mechanisms underlying the vascular actions of Ang 1-7 and Ang 1-8 in DM will facilitate the development of tailored therapeutic interventions aimed at preserving vascular health and preventing cardiovascular complications.
Collapse
Affiliation(s)
- Nazar M Shareef Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
| | - Almas M R Mahmud
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ismail M Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
61
|
Cheung YWS, Nam SE, Fairlie GMJ, Scheu K, Bui JM, Shariati HR, Gsponer J, Yip CK. Structure of the human autophagy factor EPG5 and the molecular basis of its conserved mode of interaction with Atg8-family proteins. Autophagy 2025; 21:1173-1191. [PMID: 39809444 PMCID: PMC12087653 DOI: 10.1080/15548627.2024.2447213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
The multi-step macroautophagy/autophagy process ends with the cargo-laden autophagosome fusing with the lysosome to deliver the materials to be degraded. The metazoan-specific autophagy factor EPG5 plays a crucial role in this step by enforcing fusion specificity and preventing mistargeting. How EPG5 exerts its critical function and how its deficiency leads to diverse phenotypes of the rare multi-system disorder Vici syndrome are not fully understood. Here, we report the first structure of human EPG5 (HsEPG5) determined by cryo-EM and AlphaFold2 modeling. Our structure revealed that HsEPG5 is constructed from helical bundles analogous to tethering factors in membrane trafficking pathways but contains a unique protruding thumb domain positioned adjacent to the atypical tandem LIR motifs involved in interaction with the GABARAP subfamily of Atg8-family proteins. Our NMR spectroscopic, molecular dynamics simulations and AlphaFold modeling studies showed that the HsEPG5 tandem LIR motifs only bind the canonical LIR docking site (LDS) on GABARAP without engaging in multivalent interaction. Our co-immunoprecipitation analysis further indicated that full-length HsEPG5-GABARAP interaction is mediated primarily by LIR1. Finally, our biochemical affinity isolation, X-ray crystallographic analysis, affinity measurement, and AlphaFold modeling demonstrated that this mode of binding is observed between Caenorhabditis elegans EPG-5 and its Atg8-family proteins LGG-1 and LGG-2. Collectively our work generated novel insights into the structural properties of EPG5 and how it potentially engages with the autophagosome to confer fusion specificity.ABBREVIATIONS: ATG: autophagy related; CSP: chemical shift perturbation; eGFP: enhanced green fluoresent protein; EM: electron microscopy; EPG5: ectopic P-granules 5 autophagy tethering factor; GST: glutathione S-transferase; HP: hydrophobic pocket; HSQC: heteronuclear single-quantum correlation; ITC: isothermal titration calorimetry; LDS: LC3 docking site; LIR: LC3-interacting region; MD: molecular dynamics; NMR: nuclear magnetic resonance; TEV: tobacco etch virus.
Collapse
Affiliation(s)
- Yiu Wing Sunny Cheung
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Sung-Eun Nam
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Gage M. J. Fairlie
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Karlton Scheu
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Jennifer M. Bui
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Hannah R. Shariati
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Jörg Gsponer
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Calvin K. Yip
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
62
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Gupta P, Sakthivel S, Jiemin N, Arul K, Tikno K, Park SH, Wu Y, Wang L, Bay BH, Ho L, Giguere V, Ghosh S, McDonnell DP, Yen PM, Singh BK. ESRRA (estrogen related receptor, alpha) induces ribosomal protein RPLP1-mediated adaptive hepatic translation during prolonged starvation. Autophagy 2025; 21:1283-1297. [PMID: 39936615 PMCID: PMC12087656 DOI: 10.1080/15548627.2025.2465183] [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: 01/22/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, ESRRA (estrogen related receptor, alpha) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating Rplp1 (ribosomal protein lateral stalk subunit P1) gene expression. Overexpression or siRNA knockdown of Esrra in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome and macroautophagy/autophagy protein translation, and autophagy activity. Remarkably, we have found that ESRRA had dual functions by not only regulating transcription but also controlling adaptive translation via the ESRRA-RPLP1-lysosome-autophagy pathway during prolonged starvation.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Karine Gauthier
- Département de Biologie, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, Lyon, Cedex, France
| | - Reddemma Sandireddy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Priyanka Gupta
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Suganya Sakthivel
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Nah Jiemin
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Kabilesh Arul
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Keziah Tikno
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - Lena Ho
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, Canada
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| |
Collapse
|
63
|
Liu D, Weng S, Fu C, Guo R, Chen M, Shi B, Weng J. Autophagy in Acute Lung Injury. Cell Biochem Biophys 2025; 83:1415-1425. [PMID: 39527232 DOI: 10.1007/s12013-024-01604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is a critical condition marked by rapid-onset respiratory failure due to extensive inflammation and increased pulmonary vascular permeability, often progressing to acute respiratory distress syndrome (ARDS) with high mortality. Autophagy, a cellular degradation process essential for removing damaged organelles and proteins, plays a crucial role in regulating lung injury and repair. This review examines the protective role of autophagy in maintaining cellular function and reducing inflammation and oxidative stress in ALI. It underscores the necessity of precise regulation to fully harness the therapeutic potential of autophagy in this context. We summarize the mechanisms by which autophagy influences lung injury and repair, discuss the interplay between autophagy and apoptosis, and examine potential therapeutic strategies, including autophagy inducers, targeted autophagy signaling pathways, antioxidants, anti-inflammatory drugs, gene editing, and stem cell therapy. Understanding the role of autophagy in ALI could lead to novel interventions for improving patient outcomes and reducing mortality rates associated with this severe condition.
Collapse
Affiliation(s)
- Danjuan Liu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Shuoyun Weng
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Chunjin Fu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Rongjie Guo
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Min Chen
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Bingbing Shi
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Junting Weng
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China.
| |
Collapse
|
64
|
Zhu G, Zuo Q, Liu S, Zheng P, Zhang Y, Zhang X, Rollins JA, Liu J, Pan H. A FOX transcription factor phosphorylated for regulation of autophagy facilitates fruiting body development in Sclerotinia sclerotiorum. THE NEW PHYTOLOGIST 2025; 246:2683-2701. [PMID: 40248859 DOI: 10.1111/nph.70151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025]
Abstract
Autophagy is a recycling process by which eukaryotic cells degrade their own components, and the fruiting body (sexual structure) is a necessary structure for some plant pathogenic fungi to start the infection cycle. However, the transcriptional regulation of plant pathogenic fungal autophagy and autophagy regulating sexual reproduction remains elusive. Here, we provide the report linking autophagy transcription and fruiting body development in phytopathogenic fungi. The forkhead box transcription factor (FOX TF) SsFoxE2 in Sclerotinia sclerotiorum (Ss) binds to the promoters of ATG genes, thus promoting their transcription. SsFoxE2 is phosphorylated by AMP-activated protein kinase (AMPK) SsSnf1, and the phosphorylated SsFoxE2 interacts with (translationally controlled tumor protein) SsTctp1, leading to enhanced stability and ATG transcription activity of SsFoxE2. Importantly, the regulation of autophagy by SsFoxE2 affects the balance of the ubiquitination system and the early development of the fruiting body, which directly determines the occurrence and prevalence of plant disease. Furthermore, transcriptional binding of FOX TF to ATG gene promoters is conserved in phytopathogenic fungi. Taken together, our results bring new insights into pathogen initiation in phytopathogenic fungi and connect it to other autophagy-regulated processes in plant pathogens.
Collapse
Affiliation(s)
- Genglin Zhu
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Qi Zuo
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Sirui Liu
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Peiyi Zheng
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Yanhua Zhang
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Xianghui Zhang
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Jeffrey A Rollins
- Department of Plant Pathology, University of Florida, Gainesville, FL, 32611, USA
| | - Jinliang Liu
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Hongyu Pan
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| |
Collapse
|
65
|
Yamamoto T. Autophagic stagnation: a key mechanism in kidney disease progression linked to aging and obesity. Clin Exp Nephrol 2025; 29:711-719. [PMID: 40131605 DOI: 10.1007/s10157-025-02653-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Autophagy, a critical intracellular degradation and recycling pathway mediated by lysosomes, is essential for maintaining cellular homeostasis through the quality control of proteins and organelles. Our research focused on the role of proximal tubular autophagy in the pathophysiology of aging, obesity, and diabetes. Using a novel method to monitor autophagic flux in kidney tissue, we revealed that age-associated high basal autophagy supports mitochondrial quality control and delays kidney aging. However, an impaired ability to upregulate autophagy under additional stress accelerates kidney aging. In obesity induced by a high-fat diet, lysosomal dysfunction disrupts autophagy, leading to renal lipotoxicity. Although autophagy is initially activated to repair organelle membranes and maintain proximal tubular cell integrity, this demand overwhelms lysosomes, resulting in "autophagic stagnation" characterized by phospholipid accumulation. Similar lysosomal phospholipid accumulation was observed in renal biopsies from elderly and obese patients. We identified TFEB-mediated lysosomal exocytosis as a mechanism to alleviate lipotoxicity by expelling accumulated phospholipids. Therapeutically, interventions such as the SGLT2 inhibitor empagliflozin and eicosapentaenoic acid restore lysosomal function and autophagic activity. Based on these findings, we propose a novel disease concept, "Obesity-Related Proximal Tubulopathy." This study underscores autophagic stagnation as a key driver of kidney disease progression in aging and obesity, offering insights into the pathophysiology of kidney diseases and providing a foundation for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Box D11, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
66
|
Li J, Jia K, Wang W, Pang Y, Wang H, Hao J, Zhao D, Li F. FBXW7 mediates high glucose-induced epithelial to mesenchymal transition via KLF5 in renal tubular cells of diabetic kidney disease. Tissue Cell 2025; 94:102801. [PMID: 40010183 DOI: 10.1016/j.tice.2025.102801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
F-box and WD repeat domain-containing 7 (FBXW7) protein is known as one of the crucial components of the E3 ubiquitin ligase called the Skp1-Cullin1-F-box (SCF) complex, which regulates the degradation of a network of proteins via the ubiquitin-proteasome system. In our study, we investigated the latent impact of FBXW7 on renal tubular cells injury and its molecular mechanism in diabetic kidney disease (DKD). FBXW7 was upregulated in kidneys of diabetic mice and human renal proximal tubular cells exposed to high glucose. Again, the function of experiment found that overexpression of FBXW7 led to epithelial-mesenchymal transition (EMT) of HK2 cells, as indicated by decreased E-cadherin and increased α-smooth muscle actin (α-SMA). Knockdown of FBXW7 ameliorated high glucose-induced EMT of HK2 cells via downregulation of TGF-β1. Then, FBXW7 overexpression downregulated the stability of the KLF5 protein and promoted protein ubiquitination in normal glucose-cultured HK2 cells, which was significantly reversed by the addition of MG132, a specific proteasome inhibitor. Furthermore, overexpression of KLF5 effectively prevented FBXW7 upregulation-induced EMT in HK2 cells. Finally, chemical inhibitors or mTOR kinase dead vector to interfere the activity of mTOR effectively suppressed FBXW7 expression in HK2 cells treated with high glucose. Taken together, these above data suggest that mTOR signaling pathway-regulated FBXW7 mediates high glucose-induced EMT of renal tubular cells by affecting the stability of KLF5.
Collapse
Affiliation(s)
- Juan Li
- Department of Nephrology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Keqi Jia
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Wenjie Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Yingxue Pang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Hui Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Dong Zhao
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
| | - Fan Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Kidney Diseases, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
67
|
Huang Y, Xu W, Dong W, Chen G, Sun Y, Zeng X. Anti-diabetic effect of dicaffeoylquinic acids is associated with the modulation of gut microbiota and bile acid metabolism. J Adv Res 2025; 72:17-35. [PMID: 38969095 DOI: 10.1016/j.jare.2024.06.027] [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: 03/30/2024] [Revised: 06/06/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024] Open
Abstract
INTRODUCTION The human gut microbiome plays a pivotal role in health and disease, notably through its interaction with bile acids (BAs). BAs, synthesized in the liver, undergo transformation by the gut microbiota upon excretion into the intestine, thus influencing host metabolism. However, the potential mechanisms of dicaffeoylquinic acids (DiCQAs) from Ilex kudingcha how to modulate lipid metabolism and inflammation via gut microbiota remain unclear. OBJECTIVES AND METHODS The objectives of the present study were to investigate the regulating effects of DiCQAs on diabetes and the potential mechanisms of action. Two mice models were utilized to investigate the anti-diabetic effects of DiCQAs. Additionally, analysis of gut microbiota structure and functions was conducted concurrently with the examination of DiCQAs' impact on gut microbiota carrying the bile salt hydrolase (BSH) gene, as well as on the enterohepatic circulation of BAs and related signaling pathways. RESULTS Our findings demonstrated that DiCQAs alleviated diabetic symptoms by modulating gut microbiota carrying the BSH gene. This modulation enhanced intestinal barrier integrity, increased enterohepatic circulation of conjugated BAs, and inhibited the farnesoid X receptor-fibroblast growth factor 15 (FGF15) signaling axis in the ileum. Consequently, the protein expression of hepatic FGFR4 fibroblast growth factor receptor 4 (FGFR4) decreased, accompanied by heightened BA synthesis, reduced hepatic BA stasis, and lowered levels of hepatic and plasma cholesterol. Furthermore, DiCQAs upregulated glucolipid metabolism-related proteins in the liver and muscle, including v-akt murine thymoma viral oncogene homolog (AKT)/glycogen synthase kinase 3-beta (GSK3β) and AMP-activated protein kinase (AMPK), thereby ameliorating hyperglycemia and mitigating inflammation through the down-regulation of the MAPK signaling pathway in the diabetic group. CONCLUSION Our study elucidated the anti-diabetic effects and mechanism of DiCQAs from I. kudingcha, highlighting the potential of targeting gut microbiota, particularly Acetatifactor sp011959105 and Acetatifactor muris carrying the BSH gene, as a therapeutic strategy to attenuate FXR-FGF15 signaling and ameliorate diabetes.
Collapse
Affiliation(s)
- Yujie Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Weiqi Xu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Wei Dong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Guijie Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| |
Collapse
|
68
|
Wang W, Chen J, Bao Y, Ma W, Xie Y, Wang W, Li M, Shen K. MicroRNA sequencing analysis in pediatric patients with influenza-associated acute necrotizing encephalopathy: Potential biomarkers for early diagnosis and therapy. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 130:105734. [PMID: 40120635 DOI: 10.1016/j.meegid.2025.105734] [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: 12/04/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Acute necrotizing encephalopathy (ANE) secondary to influenza infection is characterized by fulminant neurological deterioration and a high mortality rate. The underlying mechanisms remain unclear, and specific treatments are currently lacking. Therefore, understanding the pathogenesis and identifying diagnostic and therapeutic targets for influenza-induced ANE are crucial. Peripheral blood samples were collected from two groups: influenza-infected patients without ANE (mild) and influenza infection with ANE patients (severe). Differentially expressed genes (DEG) were identified through microRNA sequencing analysis, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The expression levels of the four specific miRNAs were validated using qRT-PCR. In the severe group, 24 genes were up-regulated, and 67 genes were down-regulated compared to the mild group. The expression levels of hsa-miR-1290, hsa-miR-4657, has-miR-1231, and hsa-miR-342-3p were validated by qRT-PCR, and the levels of has-miR-4657 and hsamiR- 342-3p showed significant differences between severe and mild groups. GO analysis demonstrated that the DEGs were predominantly involved in the positive regulation of cellular processes, intracellular anatomical structure, and protein binding. KEGG pathway analysis revealed that DEGs were mainly enriched in calcium signaling pathway and axon guidance. The down-regulated hsa-miR-4657 and hsa-miR-342-3p might be associated with the development of ANE in pediatric patients with influenza by regulation of calcium pathways and axon guidance.
Collapse
MESH Headings
- Humans
- MicroRNAs/genetics
- Influenza, Human/complications
- Influenza, Human/genetics
- Influenza, Human/virology
- Biomarkers/blood
- Child, Preschool
- Male
- Female
- Leukoencephalitis, Acute Hemorrhagic/diagnosis
- Leukoencephalitis, Acute Hemorrhagic/genetics
- Leukoencephalitis, Acute Hemorrhagic/etiology
- Leukoencephalitis, Acute Hemorrhagic/therapy
- Leukoencephalitis, Acute Hemorrhagic/virology
- Child
- Early Diagnosis
- Infant
- Gene Ontology
- Gene Expression Profiling
- Sequence Analysis, RNA
Collapse
Affiliation(s)
- Wei Wang
- Respiratory Department, Beijing Children's Hospital, Capital Medical University, China National Clinical Research Center of Respiratory Diseases, National Center for Children's Health, Beijing, China; Department of Respiratory, Shenzhen Children's Hospital, Shenzhen, China
| | - Jiehua Chen
- Department of Respiratory, Shenzhen Children's Hospital, Shenzhen, China
| | - Yanmin Bao
- Department of Respiratory, Shenzhen Children's Hospital, Shenzhen, China
| | - Weike Ma
- Department of Critical care medicine, Shenzhen Children's Hospital, Shenzhen, China
| | - Ying Xie
- Department of Cardiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Wenjian Wang
- Department of Respiratory, Shenzhen Children's Hospital, Shenzhen, China
| | - Meng Li
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, China.
| | - Kunling Shen
- Department of Respiratory Medicine, Beijing Children's Hospital, Capital Medical University, 56 Nanlishi Road, Beijing, China.
| |
Collapse
|
69
|
Alborzi N, Maroofi A, Hafizi Barjin Z, Moradi A, Rezvani ME, Safari F. Resveratrol attenuates pressure overload-induced myocardial remodeling in ovariectomized rats by rescuing the adaptive angiogenic response. Life Sci 2025; 370:123573. [PMID: 40122333 DOI: 10.1016/j.lfs.2025.123573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Resveratrol (RES), a polyphenol with putative estrogen (E2) -like effects, is believed to counteract left ventricular hypertrophy (LVH). However, how RES exerts its protection is not well understood, particularly when prominent risk factors, such as E2 depletion and pressure overload (PO), coexist. Here, we evaluated the impact of RES and E2 on angiogenesis and LVH in rats subjected to ovariectomy (OVX) and PO. METHODS Three weeks after bilateral OVX induction, abdominal aortic banding was performed on Wistar female rats to trigger PO. The animals were treated with either RES or E2 for six weeks. Finally, the heart-to-body weight ratio (HW/BW), cell size, fibrosis, and atrial natriuretic peptide (ANP) mRNA expression were assessed. Angiogenesis was determined by evaluating vascular endothelial growth factor (VEGF) mRNA and protein expression and by CD31 immunostaining. Serum E2 levels were also measured. RESULTS OVX + PO caused more severe myocardial hypertrophy (HW/BW) and fibrosis compared with PO alone, but did not aggravate cell size and ANP mRNA expression. OVX blunted the angiogenic response to PO, with reduced VEGF expression. RES increased VEGF expression and CD31, and abrogated LVH and fibrosis. E2 treatment improved VEGF expression and fibrosis, but not to the same extent as RES. RES improved serum levels of E2 in OVX + PO rats. CONCLUSION Our findings suggest that RES limits OVX-induced exacerbation of LVH and fibrosis in a PO model, and targets systemic E2 levels and myocardial angiogenesis as underpinning protective mechanisms. Thus, RES may provide cardioprotection for post-menopausal women.
Collapse
Affiliation(s)
- Nasrin Alborzi
- Yazd Neuroendocrine Research Center, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Zeinab Hafizi Barjin
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Moradi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ebrahim Rezvani
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
70
|
Chen J, Ran P, Xu Y, Khouchani M, Li X, Jian L, Abdelmajid T, Aittahssaint N, Yang Q, Li J, Zhao L. Biomimetic multifunctional nanoparticles for improved radiotherapy and immunotherapy in cancer treatment. Mater Today Bio 2025; 32:101698. [PMID: 40225127 PMCID: PMC11986628 DOI: 10.1016/j.mtbio.2025.101698] [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: 01/19/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/15/2025] Open
Abstract
Radiotherapy represents a conventional approach in clinical cancer treatment, but suffers from insufficient DNA damage and limited tumor selectivity. Herein, bismuth oxyiodide quantum dots loaded hollow manganese dioxide (MB) nanoparticles was fabricated and subsequently wrapped with bacterial membrane vesicles (MVs) to create MB@MV nanoparticles. This biomimetic radiosensitizer is designed to enhance the efficacy of radiotherapy through a combined approach of tumor immunotherapy and oxygen delivery strategy. Upon systemic administration, MB@MV enhance tumor accumulation through specifically targeting the inflammatory milieu mediated by MVs, thereby activating dendritic cell-mediated innate immunotherapy. Concurrently, MB@MV demonstrate superior X-ray absorption, leading to effective DNA damage in tumor cells due to the high atomic number of bismuth. Notably, manganese dioxide react with the overexpressed H2O2 in the tumor microenvironment to alleviate hypoxia and fixing X-ray induced DNA damage in tumor cells, culminating in a multi-strategy approach to enhance radiotherapy sensitization. The findings from both in vitro and in vivo experiments demonstrate a significantly enhanced inhibition of tumor growth by MB@MV compared to tumors treated solely with X-ray. Overall, our multifunctional radiosensitizer MB@MV shows considerable promise in the field of tumor radiotherapy.
Collapse
Affiliation(s)
- Jiale Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, PR China
| | - Pan Ran
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Medical College, Chengdu, 610051, PR China
- Development and Regeneration Key Laboratory of Sichuan Province, School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, PR China
| | - Yizhao Xu
- Development and Regeneration Key Laboratory of Sichuan Province, School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, PR China
| | - Mouna Khouchani
- Mohammed VI University Hospital, Cadi Ayyad University, Marrakech, Morocco
| | - Xin Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, PR China
| | - Ling Jian
- Development and Regeneration Key Laboratory of Sichuan Province, School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, PR China
| | - Takoui Abdelmajid
- Mohammed VI University Hospital, Cadi Ayyad University, Marrakech, Morocco
| | - Nadia Aittahssaint
- Mohammed VI University Hospital, Cadi Ayyad University, Marrakech, Morocco
| | - Qian Yang
- Center of Scientific Research, Chengdu Medical College, Chengdu, 610500, PR China
| | - Jingyi Li
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Medical College, Chengdu, 610051, PR China
| | - Long Zhao
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Medical College, Chengdu, 610051, PR China
- Development and Regeneration Key Laboratory of Sichuan Province, School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, PR China
| |
Collapse
|
71
|
Zargar AH, Bhansali A, Majumdar A, Maheshwari A, Bhattacharyya A, Dasgupta A, Saboo BD, Sethi BK, Sanyal D, Seshadri KG, Deshpande NR, Kapoor N, Lakhani OJ, Talwalkar PG, Kalra P, Mehrotra RN, Sahay RK, Shukla R, Kant S, Das S, Agarwal SC, Phatak SR, G S, Joshi SR, Shaikh SS, Aravind SR, Goswami S, Ghosh S, Panikar VK, Mohan V. Management of metabolic dysfunction-associated steatotic liver disease (MASLD)-An expert consensus statement from Indian diabetologists' perspective. Diabetes Obes Metab 2025; 27 Suppl 4:3-20. [PMID: 40457532 DOI: 10.1111/dom.16496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 05/12/2025] [Accepted: 05/17/2025] [Indexed: 06/11/2025]
Abstract
In India, the increasing prevalence of diabetes and obesity poses a significant threat towards a surge in the incidence of metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). Concomitant with the evolving guidelines, there is a need to direct and spread awareness among practicing diabetologists to identify and screen high-risk individuals for MASLD for timely management. Its asymptomatic nature and the evolving guidelines on diagnosis have hindered the precise estimates of MASLD in the high-risk group of individuals in a clinical setting. Therefore, an expert panel of diabetologists from India convened to review, discuss and document the approach towards screening, diagnosis and management of MASLD. Serum biomarkers, simple non-invasive tools and imaging techniques could direct the risk stratification of the patients. Early lifestyle interventions including weight loss and exercise are beneficial. The pharmacological landscape of drugs directed to insulin resistance, lipid metabolism, oxidative stress, inflammation, apoptosis and fibrogenesis pathways for the management of MASLD is expanding. In summary, the consensus statements are expected to serve as a useful guide in the screening and management of MASLD in the region and to direct a well-planned study design that could enhance the scientific value of these statements.
Collapse
Affiliation(s)
| | - Anil Bhansali
- Gini Health, Mohali, India
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anirban Majumdar
- KPC Medical College, Kolkata, India
- The University of Newcastle, Newcastle, New South Wales, Australia
| | - Anuj Maheshwari
- Hind Institute of Medical Sciences, Sitapur, India
- Sri Hari Kamal Diabetes Care & Research Centre, Lucknow, India
| | | | - Arundhati Dasgupta
- Department of Endocrinology, Rudraksh Superspeciality Care Hospital, Siliguri, India
| | | | | | - Debmalya Sanyal
- The University of Newcastle, Newcastle, New South Wales, Australia
- Department of Endocrinology, KPC Medical College, Kolkata, India
- NH Rabindranath Tagore Hospital, Kolkata, India
| | | | | | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, Vellore, India
- Non-Communicable Disease Unit, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | - Pramila Kalra
- Department of Endocrinology, Ramaiah Medical College & Memorial Hospital, Bengaluru, India
| | | | - Rakesh Kumar Sahay
- Department of Endocrinology, Osmania Medical College & Osmania General Hospital, Hyderabad, India
| | - Rishi Shukla
- Department of Endocrinology, Regency Health, Kanpur, Uttar Pradesh, India
- Centre for Diabetes and Endocrine Diseases, Kanpur, India
| | - Saket Kant
- Max Super-Speciality, Shalimar Bagh and Balaji Action Medical and Cancer Institute, Delhi, India
| | - Sambit Das
- Department of Endocrinology, Kalinga Institute of Medical Sciences, Kalinga Institute of Industrial Technology University, Bhubaneswar, India
| | - Sanjay Chunilal Agarwal
- Dr Sanjay Agarwal's Aegle Clinic for Diabetes Care, Pune, India
- Department of Internal Medicine, Ruby Hall Clinic, Pune, India
- Diabetes & Medicine, Jehangir Hospital and Apollo Group of Hospitals, Pune, India
| | | | - Shanmugasundar G
- Magna Centres for Diabetes, Obesity and Endocrinology, Chennai, India
| | | | | | | | - Soumik Goswami
- Department of Endocrinology, NRS Medical College, Kolkata, India
| | - Sujoy Ghosh
- Department of Endocrinology, Institute of Post Graduate Medical Education & Research, Kolkata, India
| | - Vijay Kumar Panikar
- Department of Endocrinology and Diabetes, Lilavati Hospital and Research Centre, Mumbai, India
- Dr. Panikar's Speciality Care Centres, Mumbai, India
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation (ICMR-Collaborating Centre of Excellence) & Dr. Mohan's Diabetes Specialities Centre (IDF Centre of Excellence in Diabetes Care), Chennai, India
| |
Collapse
|
72
|
Okunlola FO, Okunlola AR, Adetuyi BO, Soliman MES, Alexiou A, Papadakis M, Fawzy MN, El-Saber Batiha G. Beyond the gut: Unraveling the multifaceted influence of microbiome on cardiovascular health. Clin Nutr ESPEN 2025; 67:71-89. [PMID: 40064239 DOI: 10.1016/j.clnesp.2025.03.002] [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: 10/09/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Cardiovascular disease is one of the leading causes of death worldwide. Even while receiving adequate pharmacological treatment for their hypertension, people are nonetheless at greater risk for cardiovascular disease. There is growing evidence that the gut microbiota may have major positive and negative effects on blood pressure and illnesses related with it as more study into this topic is conducted. Trimethylamine n-oxide (TMAO) and short-chain fatty acids (SCFA) are two major by-products of the gut microbiota. TMAO is involved in the formation of other coronary artery diseases, including atherosclerosis and hypertension, while SCFAs play an important role in controlling blood pressure. Numerous investigations have confirmed the established link between dietary salt intake and hypertension. Reducing sodium in the diet is linked to lower rates of cardiovascular disease morbidity and mortality as well as lower rates of blood pressure and hypertension. In both human and animal research, high salt diets increase local and systemic tissue inflammation and compromise gut architecture. Given that the gut microbiota constantly interacts with the immune system and is required for the correct maturation of immune cells, it is scientifically conceivable that it mediates the inflammatory response. This review highlights the therapeutic possibilities for focusing on intestinal microbiomes as well as the potential functions of the gut microbiota and its metabolites in the development of hypertension.
Collapse
Affiliation(s)
- Felix Oladele Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Abimbola Rafiat Okunlola
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Babatunde Oluwafemi Adetuyi
- Department of Natural Sciences (Biochemistry Option), Faculty of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Nigeria.
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt.
| |
Collapse
|
73
|
Hu Q, Chen H, Lan J, Chen Y, Liu Z, Xiong Y, Zhou W, Zhong Z, Ye Q. KLF10 Induced by Hypothermic Machine Perfusion Alleviates Renal Inflammation Through BIRC2 /Noncanonical NF-κB Pathway. Transplantation 2025; 109:e273-e286. [PMID: 39716345 DOI: 10.1097/tp.0000000000005314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
BACKGROUND Hypothermic machine perfusion (HMP) is becoming the main preservation method for donation after circulatory death (DCD) kidneys. It can provide continuous flow and form shear stress (SS) upon endothelial cells (ECs), thereby regulating EC injury. Krüppel-like factor 10 (KLF10) has been shown to lessen vascular damage. However, how SS and KLF10 impact HMP-regulated injury is unclear. METHODS In this study, we investigated the influences of KLF10 on HMP in animal models and human renal biopsy and explored how SS affected KLF10 expression in a parallel-plate flow chamber system. Chromatin Immunoprecipitation sequencing and luciferase assay were performed to seek the target genes of KLF10. The influences of KLF10 on HMP-regulated injury were investigated by transfecting si-KLF10 adeno-associated virus serotype 9 into rat kidneys. The molecular expression was examined using immunofluorescence staining, Western blotting, and quantitative polymerase chain reaction. RESULTS Our results show KLF10 expression was augmented in human, rabbit, and rat DCD kidneys after HMP. HMP improved ECs and tubule injury and attenuated inflammation; however, the knockdown of KLF10 reversed this effect. SS regulated KLF10 expression in ECs by affecting F-actin, and KLF10 could maintain ECs homeostasis. Chromatin Immunoprecipitation sequencing and luciferase assay revealed that baculoviral inhibitor of apoptosis protein repeat-containing 2 (BIRC2) is a target gene of KLF10. Furthermore, BIRC2 linked to nuclear factor kappa B (NF-κB)-inducing kinase, induced NF-κB)-inducing kinase ubiquitination, and resulted in inhibiting the noncanonical NF-κB pathway. CONCLUSIONS SS can mediate KLF10 expression, whereas HMP can protect against warm ischemic injury by reducing inflammation via KLF10/BIRC2/noncanonical NF-κB pathway. Therefore, KLF10 might be a novel target for improving DCD kidney quality.
Collapse
Affiliation(s)
- Qianchao Hu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Hao Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Jia'nan Lan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Yiwen Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
- The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| |
Collapse
|
74
|
Guo Z, He L, Wang W, Tian S, Lin R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett 2025; 619:217643. [PMID: 40112906 DOI: 10.1016/j.canlet.2025.217643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Fucosyltransferase 2 (FUT2) is an enzyme that adds fucose to proteins or lipids via α-1,2-fucosylation in the intestinal mucosa. While FUT2 deficiency is linked to increased susceptibility to inflammatory bowel disease (IBD), its role in colorectal cancer (CRC) is unclear, and the molecular mechanisms involved remain largely unknown. We established an azoxymethane (AOM) and dextran sulfate sodium (DSS) model to induce CRC. FUT2 expression was assessed in human CRC tissues, AOM/DSS-induced mouse models, and CRC cell lines using qRT-PCR, western blotting, and UEA-I staining. FUT2 knockout (FUT2△IEC) mice were treated with AOM/DSS, and FUT2-overexpressing CRC cells were created to evaluate the effects of FUT2 on apoptosis in both in vitro and in vivo settings through Western blot analyses and functional assays. N-glycoproteomics, UEA-I chromatography, and co-immunoprecipitation were utilized to identify regulatory mechanisms and target fucosylated proteins. FUT2 expression and α-1,2-fucosylation were significantly decreased in CRC. FUT2 deficiency worsened AOM/DSS-induced CRC and reduced tumor apoptosis, while FUT2 overexpression induced apoptosis and inhibited proliferation in CRC cells and xenografts. Mechanistically, FUT2 appears to suppress autophagy by impairing lysosomal function and directly targeting and fucosylating LAMP1, contributing to lysosomal dysfunction. Our study reveals a fucosylation-dependent antitumor mechanism of FUT2 in CRC, suggesting potential therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingnan He
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New Area, Shanghai, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Tian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
75
|
Dong X, He Y, Hu X, Wu J, Ye F, Wang X, Zhao Y, Dan G, Zhao J, Tang H, Lu X, Sai Y, Zou Z, Chen M. Targeting LINC00707 by vitamin D3 attenuates nitrogen mustard-caused dermal toxicity through inhibiting ferroptosis. Redox Biol 2025; 83:103628. [PMID: 40245702 DOI: 10.1016/j.redox.2025.103628] [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: 03/22/2025] [Accepted: 04/05/2025] [Indexed: 04/19/2025] Open
Abstract
Nitrogen mustard (NM) causes severe skin injury that is lack of effective and targeted therapies. Vitamin D3 (VD3) emerges as a promising treatment option for NM-caused dermal toxicity; however, the underlying mechanisms are currently unclear. Herein, we identified that NM markedly promoted ferroptosis by measurement of decreased cell viability, glutathione, glutathione peroxidase 4 and solute carrier family 7 member 11 levels, and increased ROS, lipid ROS, iron/Fe2+ and malondialdehyde contents in vitro and in vivo. Ferrostin-1 (Fer-1, a ferroptosis inhibitor) attenuated NM-caused cell death in keratinocytes. Meanwhile, NM significantly inhibited phosphorylation of AKT1 and glycogen synthase kinase 3β (GSK3β) and nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, and increased LINC00707 expression. Furthermore, NM-induced ferroptosis in keratinocytes was abolished by treatment with agonists of Nrf2 (tBHQ) and AKT1 (SC79), the inhibitor of GSK3β (AR-A014418), Nrf2 overexpression or LINC00707 knockdown. Mechanistically, LINC00707 directly bound with the protein kinase domain of AKT1 and suppressed its phosphorylation and activated GSK3β thereby inactivating Nrf2, subsequently inducing ferroptosis and cell death in NM-treated keratinocytes. Moreover, VD3 notably suppressed LINC00707 expression, activated AKT1 and inactivated GSK3β, increased Nrf2 nuclear translocation and inhibited ferroptosis and cytotoxicity induced by NM in vitro and in vivo. The protective effects of VD3 against NM-caused dermal toxicity were blocked by erastin (a ferroptosis inducer), Nrf2 siRNA, LINC00707 overexpression and were enhanced by LINC00707 knockdown and Fer-1 in vitro and in vivo. In conclusion, VD3 ameliorated NM-caused dermal toxicity by inhibiting ferroptosis, which was partially mediated through the LINC00707-AKT1-GSK3β-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Xunhu Dong
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Ying He
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiaofeng Hu
- Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Jie Wu
- Department of Tropical Medicine, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Feng Ye
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Xiaogang Wang
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Yuanpeng Zhao
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Guorong Dan
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jiqing Zhao
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - He Tang
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Xiaolu Lu
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Yan Sai
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, China.
| | - Zhongmin Zou
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| | - Mingliang Chen
- Institute of Toxicology, School of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, China.
| |
Collapse
|
76
|
Wan Y, Zhang L, Wang L, Zhang Y, Gao F. High-altitude chronic hypoxia prevents myocardial dysfunction in experimental model of type 2 diabetes. Arch Biochem Biophys 2025; 768:110371. [PMID: 40086566 DOI: 10.1016/j.abb.2025.110371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND High-altitude chronic hypoxia (CHH) has a favorable impact on the lower prevalence of diabetes together with the better glucose tolerance. However, whether it prevents diabetic cardiomyopathy remains unclear. This study aimed to investigate the effects of CHH on left ventricular (LV) function in experimental model of type 2 diabetes. METHODS Sprague-Dawley rats were randomly divided into control (altitude 500 m), DM (diabetes mellitus and altitude 500 m), CHH (altitude 4250 m and non-diabetic for 2 weeks), CHH-DM2 (altitude 4250 m and DM for 2 weeks), and CHH-DM8 (altitude 4250 m and DM for 8 weeks) groups. The experimental model of type 2 diabetes was induced by a high-fat diet plus low-dose streptozotocin (35 mg/kg, intraperitoneal) after fasted overnight. Left ventricular cardiac function and global myocardial strain were evaluated at 2, and 8 weeks by 7.0 T cardiovascular magnetic resonance. Subsequently, biochemical indices, histological evaluation, and levels of hypoxia-induced factor (HIF)-1α were assessed. RESULTS Left ventricular ejection fraction (LVEF), global longitudinal (GLS), circumferential (GCS), and radial (GRS) strains significantly decreased in the DM group compared with the controls. However, these abnormalities in DM rats were significantly prevented in the CHH-DM2 group, and were further improved in CHH-DM8 group. Mechanistically, prolonged CHH at high altitude further reduced cardiac apoptosis, and oxidative stress, and increased autophagy, and the expression of HIF-1α in diabetic myocardial tissue. CONCLUSIONS CHH exerted cardioprotective effects by improving LV function, increasing myocardial strain and attenuating cardiac hypertrophy in type 2 diabetic rats, likely through reducing apoptosis and oxidative stress, activating autophagy and HIF-1α signaling in diabetic rats.
Collapse
Affiliation(s)
- Yixuan Wan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lisha Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Wang
- Molecular Imaging Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - FaBao Gao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
77
|
Gardener H, Bowen J, Callan SP. Heavy metals and phthalate contamination in prenatal vitamins and folic acid supplements. ENVIRONMENTAL RESEARCH 2025; 274:121255. [PMID: 40020868 DOI: 10.1016/j.envres.2025.121255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/03/2025]
Abstract
OBJECTIVE The goal is to characterize the contamination of prenatal vitamins and folate/folic acid supplements with lead, cadmium, and phthalates. METHODS The sample included 156 commercially available prenatal vitamins, 19 folate/folic acid supplements, and nine prescription prenatal supplements. Lead and cadmium were measured by inductively coupled plasma-mass spectrometry, and phthalates by liquid chromatography-tandem mass spectrometry, and quantified as μg/daily serving. Distributions of lead, cadmium, and phthalates were examined across products, as well as the proportion exceeding the California proposition 65 threshold for daily lead consumption (0.5 μg). RESULTS Lead exceeded the limit of quantification (LOQ) in 83% of commercially available prenatal vitamin samples (15% > 0.5 μg/serving), cadmium in 73%, DEHP in 25%, and DBP in 13%. Product characteristics associated with increased lead and cadmium contamination included calcium and iron doses, and being a caplet, capsule, or tablet. Lead and cadmium exceeded the LOQ in 7/9 prescription prenatals (33% > 0.5 μg/serving lead). Heavy metal and phthalate contamination was lower in folate/folic acid supplements. CONCLUSIONS Clear and enforceable regulations regarding frequent testing and restriction of lead and cadmium contamination in prenatal vitamins are needed.
Collapse
Affiliation(s)
- Hannah Gardener
- University of Miami, Miller School of Medicine, Department of Neurology, Miami, Florida, USA.
| | | | | |
Collapse
|
78
|
Li Y, Pan L, Zhao Q, Xiong H, Fang X, Guo X, Wang Y, Su X, Liu P, Hao J, Wang L, Wang X, Dong Q, Li Z, Jin F. Tanshinone IIA + Osthole alleviates ferroptosis in LPS-induced acute lung injury by Keap1-Nrf2/HO-1 pathway. Microb Pathog 2025; 203:107347. [PMID: 40147558 DOI: 10.1016/j.micpath.2025.107347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Acute lung injury (ALI) is associated with a high mortality rate and requires effective treatment. Tanshinone IIA (T) and Osthole (O) exhibit anti-inflammatory effects and have been used to protect against lipopolysaccharide (LPS)-induced lung injury in mice. However, the combined effects of T and O on lung injury protection and their potential protective mechanisms have not been studied. OBJECTIVE To assess the protective effects of TO on LPS-induced ALI in mice and BEAS-2B cell injury and to investigate the potential mechanisms underlying these protective effects. METHODS Models of ALI induced by LPS were established. The assessment encompassed the viability of BEAS-2B cells, cell count, myeloperoxidase (MPO) activity, protein content, as well as IL-6 and TNF-a levels in bronchoalveolar lavage fluid (BALF). Additionally, malondialdehyde (MDA), reactive oxygen species (ROS), and glutathione (GSH) levels in mouse lung tissue were measured. The effects of TO were assessed using immunofluorescence (IF), immunohistochemistry (IHC), Western Blot (WB), RT-PCR, and ELISA. Statistical analysis involved one-way ANOVA and t-test. RESULTS TO administration led to a significant reduction in lung edema (W/D), MDA, ROS, GSH, and superoxide dismutase (SOD) levels compared to the individual T or O groups, alleviating LPS-induced ALI. TO also significantly attenuated lung tissue damage, reduced inflammatory response, decreased Fe2+ and 4-HNE levels, and increased GPX4, SLC7A11, and Nrf2 gene expression in mice. Ultimately, TO alleviated ferroptosis in LPS-induced ALI by activating Nrf2 expression, and no markedly adverse reactions were observed. CONCLUSION TO alleviates LPS-induced ALI and effectively treats against LPS-induced ALI.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Qi Zhao
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Huanqing Xiong
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xiaoan Fang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xian Guo
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Ying Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xiaorong Su
- Department of Endocrinology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Pan Liu
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Jing Hao
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Lei Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Xinqun Wang
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China
| | - Quandi Dong
- General Surgery Department, 947 Army Hospital, China.
| | - Zhichao Li
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China.
| | - Faguang Jin
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710038, China.
| |
Collapse
|
79
|
Scardino B, Dicharry D, Agrawal A, Xing D, Bhuiyan MMR, Bhuiyan MS, Rom O, Conrad SA, Vanchiere JA, Orr AW, Kevil CG, Bhuiyan MAN. Cumulative environmental exposures and metabolic syndrome: A study of heavy metals and volatile organic compounds. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118238. [PMID: 40286740 DOI: 10.1016/j.ecoenv.2025.118238] [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: 01/20/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Metabolic Syndrome (MetS), a condition affecting over one-third of the U.S. population, heightens the risk of cardiovascular disease, Type 2 diabetes, and premature mortality. While individual links between heavy metals (HM), volatile organic compounds (VOC), and MetS have been established, the impact when these environmental toxins are combined remains unclear and unexplored. This study investigates how simultaneous exposure to HMs and VOCs influences the risk of MetS. METHODS Weighted Quantile Sum regression and Bayesian kernel Machine Regression were performed on data from 6603 participants in the National Health and Nutrition Examination Survey (2011-2020) to determine the impact of HMs and VOCs detected in urine on MetS. Further analyses were performed for individuals placed in subgroups based on age, sex, race/ethnicity, and monthly poverty level index. RESULTS The analyses reveal that combined exposure to HMs and VOCs is associated with an increased risk of MetS; in particular, exposure to cadmium, tin, N-acetyl-S-(N-methyl carbamoyl)-L-cysteine, and N-acetyl-S-(2-carboxyethyl)-L-cysteine significantly elevates the risk of developing MetS. Younger adults (18-50 years), men, Hispanics and non-Hispanic whites, and those with a monthly poverty index > 1.3 (higher socioeconomic status) emerged as the most vulnerable groups. CONCLUSION These findings emphasize an urgent need to address and tackle the cumulative impact of environmental toxins through a shift in public health efforts to go beyond investigating isolated exposures to address real-world chemical exposures. By understanding these cumulative risks, we can begin to mitigate them and pave the way for more effective interventions, especially for at-risk populations.
Collapse
Affiliation(s)
- Brooke Scardino
- Department of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Destyn Dicharry
- Department of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Akshat Agrawal
- Department of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Diensn Xing
- Department of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | | | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Steven A Conrad
- Department of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; Department of Pediatrics, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - John A Vanchiere
- Department of Pediatrics, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Mohammad Alfrad Nobel Bhuiyan
- Department of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA.
| |
Collapse
|
80
|
Alarcón-Veleiro C, López-Calvo I, Berjawi L, Lucio-Gallego S, Mato-Basalo R, Quindos-Varela M, Lesta-Mellid R, Santamarina-Caínzos I, Varela-Rodríguez S, Fraga M, Quintela M, Vizoso-Vázquez A, Arufe MC, Fafián-Labora J. Ferroptosis: An emerging strategy for managing epithelial ovarian cancer. Biomed Pharmacother 2025; 187:118065. [PMID: 40306179 DOI: 10.1016/j.biopha.2025.118065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Ferroptosis is a regulated form of cell death characterised by iron-dependent lipid peroxidation, a process intricately linked to cellular redox homeostasis. This form of cell death is induced by the accumulation of intracellular iron and the subsequent generation of reactive oxygen species (ROS), which leads to lipid peroxidation and ultimately cell death. Ferroptosis is distinct from traditional forms of cell death, such as apoptosis, and holds significant therapeutic potential, particularly in cancers harboring rat sarcoma virus (RAS) mutations, such as epithelial ovarian cancer (EOC). EOC is notoriously resistant to conventional therapies and is associated with a poor prognosis. In this review, we examine recent progress in the understanding of ferroptosis, with a particular focus on its redox biology and the complex regulatory networks involved. We also propose a novel classification system for ferroptosis modulators, grouping them into six categories (I, II, III, IV, V and VI) based on their mechanisms of action and their roles in modulating cellular redox status. By refining these categories, we aim to provide deeper insights into the role of ferroptosis in cancer biology, especially in EOC, and to identify potential therapeutic avenues. We propose that further investigation of ferroptosis in the context of redox biology could reveal novel biomarkers and therapeutic targets, offering promising strategies to overcome resistance mechanisms and improve clinical outcomes for patients with EOC and other treatment-resistant cancers.
Collapse
Affiliation(s)
- C Alarcón-Veleiro
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - I López-Calvo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain; Centro Interdisciplinar de Química de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Rúa as Xubias 84, A Coruña 15006, Spain
| | - L Berjawi
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - S Lucio-Gallego
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - R Mato-Basalo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - M Quindos-Varela
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - R Lesta-Mellid
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - I Santamarina-Caínzos
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - S Varela-Rodríguez
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - M Fraga
- Department of Anatomical Pathology, University Hospital Complex A Coruña, As Xubias 84, A Coruña 15006, Spain
| | - M Quintela
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - A Vizoso-Vázquez
- Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain
| | - M C Arufe
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| | - J Fafián-Labora
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| |
Collapse
|
81
|
Jiang J, Hu S, Hu K, Xiao L, Lin J, Chen Y, Zhang D, Ou Y, Zhang J, Yuan L, Wang W, Yu P. Novel impact of metal ion-induced cell death on diabetic cardiomyopathy pathogenesis and therapy. Apoptosis 2025; 30:1152-1181. [PMID: 40042744 DOI: 10.1007/s10495-025-02090-4] [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] [Accepted: 02/07/2025] [Indexed: 06/16/2025]
Abstract
Diabetes mellitus is a common chronic metabolic disease, with its prevalence escalating annually. Diabetic cardiomyopathy is a leading cause of mortality among diabetic patients, characterized by intricate metabolic disturbances and myocardial cell demise. Various forms of cellular death pathways including apoptosis, pyroptosis, autophagic cell death, necroptosis, ferroptosis, and entosis have been identified in diabetic cardiomyopathy. Inhibiting myocardial cell death pathways has shown promise in mitigating diabetic cardiomyopathy progression. However, there are still gaps in understanding the role of metal ions in diabetic cardiomyopathy pathogenesis. Recent research endeavors have found that iron, copper, zinc, calcium, manganese and other metal elements related to cell death play an intricate and critical role in the pathogenesis and progression of diabetic cardiomyopathy. Notably, many animal studies have shown that the development and progression of diabetic cardiomyopathy can be alleviated by inhibiting the cell death process induced by these metal ions. Therefore, we review the molecular mechanisms underlying the death of various metal ions and the potential pathophysiological roles they play in diabetic cardiomyopathy. In addition, the value of these metal ions in the treatment of diabetic cardiomyopathy is also described.
Collapse
Affiliation(s)
- Jingjing Jiang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, China
| | - Shengnan Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Kaibo Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Leyang Xiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Jitao Lin
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999007, Hong Kong
| | - Yangliu Ou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Linhui Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Wenting Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Hainan University, Haikou, 570311, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
82
|
Cho S, Tam E, Nguyen K, Lei Y, Fillebeen C, Pantopoulos K, Sung HK, Sweeney G. ω-6 PUFA-enriched membrane phospholipid composition of cardiomyocytes increases the susceptibility to iron-induced ferroptosis and inflammation. Apoptosis 2025; 30:1614-1627. [PMID: 40381101 DOI: 10.1007/s10495-025-02121-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/19/2025]
Abstract
Ferroptosis is an attractive therapeutic target in cardiometabolic disease (CMD); however, its contribution to myocardial damage requires further elucidation. This study was designed to examine whether altered phospholipid composition in cardiomyocytes enhanced ferroptosis susceptibility, and the underlying mechanisms. Human iPSC-derived cardiomyocytes and H9c2 cells were used to study iron-induced lipid peroxidation, cell death, and inflammation after exposure to different types of fatty acids. Lipidomic analysis was performed using LC/MS to assess changes in phospholipid composition, with a focus on ω-6 PUFA-containing phospholipids. Cellular and mitochondrial lipid peroxidation, sterile inflammation, and cell death were evaluated. Additionally, the release of damage-associated molecular patterns (DAMPs) and macrophage responses, including STING and type I interferon (IFN-I) signaling, were investigated. LC/MS lipidomic analysis indicated that treating cells with arachidonic acid (AA) elevated ω-6 PUFA-containing phospholipids, particularly phosphatidylethanolamines (PE) and phosphatidylcholines (PC). This significantly increased susceptibility to iron-induced total cellular as well as mitochondrial lipid peroxidation. Subsequently, increased release of mitochondrial DNA to cytosol was detected, resulting in both sterile inflammation and subsequent cell death. Furthermore, iron-induced release of one or more damage associated molecular patterns (DAMP) from AA-treated cells that induced crosstalk with macrophages eliciting a STING and type I interferon (IFN-I) response. These results indicate that cardiomyocytes enriched with ω-6 PUFA-containing phospholipids are more susceptible to lipid peroxidation, underscoring ferroptosis as a critical factor in myocardial damage associated with CMD.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
| | - Eddie Tam
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
| | - Khang Nguyen
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
| | - Yubin Lei
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada
| | - Carine Fillebeen
- Jewish General Hospital and Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Kostas Pantopoulos
- Jewish General Hospital and Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Hye Kyoung Sung
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada.
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, M3J1P3, Canada.
| |
Collapse
|
83
|
Zhang J, Tao J, Zhou Z, Pei W, Xiao Y, Guo Y, Gao J, Jiang C, Dai L, Zhang G, Tan C. Current research on mitochondria‑associated membranes in cardiovascular diseases (Review). Mol Med Rep 2025; 31:141. [PMID: 40183396 PMCID: PMC11976516 DOI: 10.3892/mmr.2025.13506] [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: 10/14/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025] Open
Abstract
The present study aimed to explore the role of mitochondria‑associated membranes (MAMs) as a key interface between mitochondria and the endoplasmic reticulum (ER) and to evaluate their importance in maintaining the physiological functions of these two organelles. MAMs not only act as a structural bridge between mitochondria and the ER but also widely participate in the regulation of mitochondrial biosynthesis and function, Ca2+ signal transduction, lipid metabolism, oxidative stress response and autophagy. In addition, the specific protein composition of MAMs is increasingly being recognized as having a profound impact on their function, and these proteins play a central role in regulating intercellular communication. Recently, the scientific community has accumulated a large amount of evidence supporting MAMs as potential targets for cardiovascular disease treatment. The present review focuses on the fine structure and multifunctional properties of MAMs and their mechanisms in the occurrence and development of cardiovascular diseases. The goal is to explore the mechanism of MAMs, therapeutic intervention points directly related to cardiovascular diseases, and feasibility of incorporating MAMs into the diagnostic strategy and treatment plan of cardiovascular diseases to provide novel insights and theoretical support for clinical practice in this field. MAMs have great potential as therapeutic targets for various cardiovascular diseases. This finding not only deepens the understanding of the interaction between organelles but also opens up a promising research path for the development of new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiaheng Zhang
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Jing Tao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Zijuan Zhou
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wanjuan Pei
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yili Xiao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yanghongxu Guo
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jian Gao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Chenyv Jiang
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Ling Dai
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Guomin Zhang
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Chao Tan
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Inherit Workroom of Medical Master Professor Xiong Ji-bo's Experiences, First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
84
|
Clain J, Couret D, Bringart M, Meilhac O, Lefebvre d’Hellencourt C, Diotel N. Effect of metabolic disorders on reactive gliosis and glial scarring at the early subacute phase of stroke in a mouse model of diabetes and obesity. IBRO Neurosci Rep 2025; 18:16-30. [PMID: 39816479 PMCID: PMC11733059 DOI: 10.1016/j.ibneur.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025] Open
Abstract
It is well recognized that type II Diabetes (T2D) and overweight/obesity are established risk factors for stroke, worsening also their consequences. However, the underlying mechanisms by which these disorders aggravate outcomes are not yet clear limiting the therapeutic opportunities. To fill this gap, we characterized, for the first time, the effects of T2D and obesity on the brain repair mechanisms occurring 7 days after stroke, notably glial scarring. In the present study, by performing a 30-minute middle cerebral artery occlusion (MCAO) on db/db (obese diabetics mice) and db/+ (controls) mice, we demonstrated that obese and diabetic mice displayed larger lesions (i.e. increased infarct volume, ischemic core, apoptotic cell number) and worsened neurological outcomes compared to their control littermates. We then investigated the formation of the glial scar in control and db/db mice 7 days post-stroke. Our observations argue in favor of a stronger and more persistent activation of astrocytes and microglia in db/db mice. Furthermore, an increased deposition of extracellular matrix (ECM) was observed in db/db vs control mice (i.e. chondroitin sulfate proteoglycan and collagen type IV). Consequently, we demonstrated for the first time that the db/db status is associated with increased astrocytic and microglial activation 7 days after stroke and resulted in higher deposition of ECM within the damaged area. Interestingly, the injury-induced neurogenesis appeared stronger in db/db as shown by the labeling of migrating neuroblast. This increase appeared correlated to the larger size of lesion. It nevertheless raises the question of the functional integration of the new neurons in db/db mice given the observed dense ECM, known to be repulsive for neuronal migration. Carefully limiting glial scar formation after stroke represents a promising area of research for reducing neuronal loss and limiting disability in diabetic/obese patients.
Collapse
Affiliation(s)
- Julien Clain
- Université de la Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre 97410, France
| | - David Couret
- Université de la Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre 97410, France
- CHU de La Réunion, Saint-Pierre 97410, France
| | - Matthieu Bringart
- Université de la Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre 97410, France
| | - Olivier Meilhac
- Université de la Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre 97410, France
- CHU de La Réunion, Saint-Pierre 97410, France
| | - Christian Lefebvre d’Hellencourt
- Université de la Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre 97410, France
| | - Nicolas Diotel
- Université de la Réunion, INSERM, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre 97410, France
| |
Collapse
|
85
|
Kiani P, Khodadadi ES, Nikdasti A, Yarahmadi S, Gheibi M, Yousefi Z, Ehtiati S, Yahyazadeh S, Shafiee SM, Taghizadeh M, Igder S, Khatami SH, Karima S, Vakili O, Pourfarzam M. Autophagy and the peroxisome proliferator-activated receptor signaling pathway: A molecular ballet in lipid metabolism and homeostasis. Mol Cell Biochem 2025; 480:3477-3499. [PMID: 39891864 DOI: 10.1007/s11010-025-05207-0] [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: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
Lipids, which are indispensable for cellular architecture and energy storage, predominantly consist of triglycerides (TGs), phospholipids, cholesterol, and their derivatives. These hydrophobic entities are housed within dynamic lipid droplets (LDs), which expand and contract in response to nutrient availability. Historically perceived as a cellular waste disposal mechanism, autophagy has now been recognized as a crucial regulator of metabolism. Within this framework, lipophagy, the selective degradation of LDs, plays a fundamental role in maintaining lipid homeostasis. Dysregulated lipid metabolism and autophagy are frequently associated with metabolic disorders such as obesity and atherosclerosis. In this context, peroxisome proliferator-activated receptors (PPARs), particularly PPAR-γ, serve as intracellular lipid sensors and master regulators of gene expression. Their regulatory influence extends to both autophagy and lipid metabolism, indicating a complex interplay between these processes. This review explores the hypothesis that PPARs may directly modulate autophagy within the realm of lipid metabolism, thereby contributing to the pathogenesis of metabolic diseases. By elucidating the underlying molecular mechanisms, we aim to provide a comprehensive understanding of the intricate regulatory network that connects PPARs, autophagy, and lipid homeostasis. The crosstalk between PPARs and other signaling pathways underscores the complexity of their regulatory functions and the potential for therapeutic interventions targeting these pathways. The intricate relationships among PPARs, autophagy, and lipid metabolism represent a pivotal area of research with significant implications for understanding and treating metabolic disorders.
Collapse
Affiliation(s)
- Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elaheh Sadat Khodadadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122, Padova, Italy
| | - Ali Nikdasti
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Sahar Yarahmadi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mobina Gheibi
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeynab Yousefi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Motahareh Taghizadeh
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
86
|
Askarizadeh F, Butler AE, Kesharwani P, Sahebkar A. Regulatory effect of curcumin on CD40:CD40L interaction and therapeutic implications. Food Chem Toxicol 2025; 200:115369. [PMID: 40043936 DOI: 10.1016/j.fct.2025.115369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/24/2025] [Accepted: 03/02/2025] [Indexed: 04/21/2025]
Abstract
Natural compounds have garnered significant attention as potential therapeutic agents due to their inherent properties. Their notable qualities, including safety, efficacy, favorable pharmacokinetic properties, and heightened effectiveness against certain diseases, particularly inflammatory conditions, make them particularly appealing. Among these compounds, curcumin has attracted considerable interest for its unique therapeutic properties and has therefore been extensively studied as a potential therapeutic agent for treating various diseases. Curcumin exhibits diverse anti-inflammatory, antioxidant, and antimicrobial effects. Curcumin's immune system regulatory ability has made it a promising compound for treatment of various inflammatory diseases, such as psoriasis, atherosclerosis, asthma, colitis, IBD, and arthritis. Among the signaling pathways implicated in these conditions, the CD40 receptor together with its ligand, CD40L, are recognized as central players. Studies have demonstrated that the interaction between CD40 and CD40L interaction acts as the primary mediator of the immune response in inflammatory diseases. Numerous studies have explored the impact of curcumin on the CD40:CD40L pathway, highlighting its regulatory effects on this inflammatory pathway and its potential therapeutic use in related inflammatory conditions. In this review, we will consider the evidence concerning curcumin's modulatory effects in inflammatory disease and its potential therapeutic role in regulating the CD40:CD40L pathway.
Collapse
Affiliation(s)
- Fatemeh Askarizadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
87
|
Alcover S, López S, Ramos-Regalado L, Muñoz-García N, Gallinat A, Suades R, Badimon L, Vilahur G. Cardioprotection During Myocardial Infarction in Diabetic Cardiomyopathy. Diabetes 2025; 74:1021-1032. [PMID: 40080393 PMCID: PMC12097457 DOI: 10.2337/db24-0510] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Patients with diabetes are at an increased risk of diabetic cardiomyopathy (DCM) and acute myocardial infarction (AMI). Protecting the heart against AMI is more challenging in DCM than in nondiabetic hearts. We investigated whether intravenous (i.v.) atorvastatin administration during AMI exerts cardioprotection in DCM as seen in nondiabetic hearts. Sprague-Dawley rats were divided into streptozotocin-induced DCM and normoglycemic control groups. Our model of DCM rats exhibited interstitial fibrosis and cardiac dysfunction at 5 weeks. At this time point, all animals underwent AMI induction (coronary ligation for 45 min), receiving i.v. atorvastatin or vehicle during ischemia. Animals were reperfused and sacrificed 24 h later for myocardial infarct size analysis and cardiac tissue sampling. Echocardiography was performed. DCM vehicle rats had larger infarcts than normoglycemic vehicle-treated animals at a comparable area-at-risk. Intravenous atorvastatin reduced infarct size and preserved systolic function in both groups. Compared with vehicle animals, i.v. atorvastatin inhibited RhoA membrane translocation, induced AMPK phosphorylation, prevented apoptosis execution, and improved cardiac remodelling in the infarcted heart of both groups, whereas innate immune cell infiltration was further reduced in i.v. atorvastatin-treated DCM animals. The proven cardioprotective effectiveness of this i.v. statin formulation in the presence of DCM warrants its further development into a clinically therapeutic option. ARTICLE HIGHLIGHTS Diabetic cardiomyopathy (DCM) significantly increases the risk of acute myocardial infarction and attenuates or abolishes the cardioprotective effects of several therapeutic approaches. Whether intravenous atorvastatin administration during ongoing acute myocardial infarction retains its cardioprotective potential in the presence of DCM was investigated. Intravenous atorvastatin during ischemia reduces infarct size and preserves cardiac function in DCM rats. The efficacy of this intravenous statin formulation in DCM supports its development as a viable therapeutic option for clinical use.
Collapse
Affiliation(s)
| | - Sergi López
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | | | | | - Alex Gallinat
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | - Rosa Suades
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
| | - Lina Badimon
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
- CiberCV, Institute of Health Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Sant Pau Research Institute (IR SANT PAU), Barcelona, Spain
- CiberCV, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
88
|
Li X, Qu S. Novel insights into the central protective role of ACE2 in diabetic cardiomyopathy: from underlying signaling pathways to therapeutic perspectives. Mol Cell Biochem 2025; 480:3535-3551. [PMID: 39928210 DOI: 10.1007/s11010-024-05196-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a cardiac complication specific to individuals with diabetes. It is defined as abnormalities of myocardial structure and function in diabetic patients who do not exhibit any obvious coronary artery disease, hypertensive heart disease, valvular heart disease, or inherited cardiomyopathy. A significant cardiovascular protective factor identified recently is angiotensin-converting enzyme 2 (ACE2), which is a rising star in the renin angiotensin system (RAS) and is responsible for the onset and progression of DCM. Nonetheless, there is not a comprehensive review outlining ACE2's effect on DCM. From the perspective of the pathogenesis of DCM, this review summarizes the myocardial protective role of ACE2 in the aspects of alleviating myocardial structure and dysfunction, correcting energy metabolism disorders, and restoring vascular function. Concurrently, we propose the connections between ACE2 and underlying signaling pathways, including ADAM17, Apelin/APJ, and Nrf2. Additionally, we highlight ACE2-related pharmaceutical treatment options and clinical application prospects for preventing and managing DCM. Further and underlying research is extensively required to completely comprehend the principal pathophysiological mechanism of DCM and the distinctive function of ACE2, switching experimental findings into clinical practice and identifying efficient therapeutic approaches.
Collapse
Affiliation(s)
- Xinyi Li
- Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hongxiang Street, Hengyang, 421001, Hunan, China.
| |
Collapse
|
89
|
Wang J, Dong J, Xu Q, Yan S, Wang H, Lei H, Ma X, Yang T, Wang K, Li Z, Wang X. Melatonin ameliorates RF-EMR-induced reproductive damage by inhibiting ferroptosis through Nrf2 pathway activation. Pathol Res Pract 2025; 270:156003. [PMID: 40344840 DOI: 10.1016/j.prp.2025.156003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
In recent years, there has been increased attention to the deleterious impacts of radiofrequency electromagnetic radiation (RF-EMR) on male reproductive ability, necessitating the exploration of effective protective measures. Melatonin has antioxidant and anti-apoptotic effects, and there is growing evidence of its benefit to the reproductive process. However, the biochemical mechanisms by which melatonin protects against reproductive damage from RF-EMR exposure are unknown. Here, we found that prolonged (8 weeks) exposure to RF-EMR [2.45 GHz; power density, 2.5 W/m2; whole-body specific absorption rate (SAR), 0.125-0.5 W/kg] induced ferroptosis and oxidative stress in testicular tissue, leading to a decrease of sperm quality in male mice. Notably, the administration of melatonin mitigated the oxidative harm to the testicles and ferroptosis caused by RF-EMR in mice. Mechanistically, melatonin could inhibit ROS production and ferroptosis by stimulating the nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling pathway through its receptors (MT1/MT2). Taken together, these results indicate that melatonin could potentially improve RF-EMR-induced reproductive damage in male mice by blocking ferroptosis through activation of the Nrf2 pathway.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Jie Dong
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Qian Xu
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China; Department of Reproductive Medicine, General Hospital of Chinese PLA Central Theater Command, Wuhan, Hubei Province, China
| | - Song Yan
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Haihui Wang
- Basic Medicine School, Air Force Medical University, No.5 Cadet Regiment, Xi'an, Shaanxi, China
| | - Hui Lei
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Xuhui Ma
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Tao Yang
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Ke Wang
- Department of Reproductive Medicine, Xi'an Gaoxin Hospital, Xi'an, Shaanxi Province, China.
| | - Zhen Li
- Department of Histology and Embryology, Air Force Medical University, Xi'an, Shaanxi Province, China.
| | - Xiaohong Wang
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
90
|
Huang C, Luo Y, Liu Y, Liu J, Chen Y, Zeng B, Liao X, Liu Y, Wang X. DNA hypermethylation-induced suppression of ALKBH5 is required for folic acid to alleviate hepatic lipid deposition by enhancing autophagy in an ATG12-dependent manner. J Nutr Biochem 2025; 140:109870. [PMID: 39993647 DOI: 10.1016/j.jnutbio.2025.109870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/08/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs when too much fat builds up in the liver. As a growing worldwide epidemic, NAFLD is strongly linked with multiple metabolic diseases including obesity, insulin resistance, and dyslipidemia. However, very few effective treatments are currently available. Folate, an essential B-group vitamin with important biological functions including DNA and RNA methylation regulation, has been shown to have a protective effect against NAFLD with its underlying mechanism remains largely unclear. Here, we show that administration of folic acid significantly improves glucose tolerance, insulin sensitivity, and dyslipidemia in high-fat diet (HFD) fed mice. Moreover, folic acid treatment significantly inhibits lipid deposition in hepatocytes both in vivo and in vitro. Mechanically, folic acid reduces the expression of m6A demethylase AlkB homolog 5 (ALKHB5) via promoter DNA hypermethylation. Decreased ALKBH5 causes increased m6A modification and increased expression of ATG12 in a demethylase activity-dependent manner, thereby promoting autophagy and preventing hepatic steatosis. Inhibition of ATG12 induced by overexpression of ALKBH5 could impair autophagy and the inhibitory effect of folic acid on lipid accumulation in hepatocytes. Together, these findings provide novel insights into understanding the protective role of folic acid in the treatment of NAFLD and suggest that folic acid may be a potential agent for combating NAFLD.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China.
| |
Collapse
|
91
|
Ghobakhloo S, Khoshakhlagh AH, Mostafaii GR, Carlsen L. Biomonitoring of metals in the blood and urine of waste recyclers from exposure to airborne fine particulate matter (PM 2.5). JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2025; 23:2. [PMID: 39583880 PMCID: PMC11582262 DOI: 10.1007/s40201-024-00924-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
This is the first systematic investigation of occupational exposure to toxic metals among waste recyclers in municipal waste recycling facilities. Concentrations of heavy metals (HMs) in the blood and urine of exposed recyclers in different jobs were compared to control groups (administrative department), identifying possible work-related and socio-demographic exposure factors. The potential relationship between HMs levels in PM2.5 and HM concentrations in the blood and urine of recyclers was studied for ten elements. Mean concentrations of HMs of recyclers were significantly higher than for the control group. Over 50% of the waste recyclers had HM levels higher than the recommended limits. The study revealed that most of the waste recyclers engaged in a minimum of three tasks, posing a challenge in establishing a correlation between specific tasks and the levels of elements monitored through biomonitoring. Co levels in blood and Fe levels in the urine of waste recyclers have a significant relationship with the increase in daily working hours. Among the variables related to the participant's demographic information, the level of education and monthly income were significantly different compared to the control group. Also, a significant correlation was found between HM levels in PM2.5 personal exposure and recyclers' urine and blood. Management controls include workflow or, in other words, alternate relocation of workers exposed to severe risks. Engineering controls such as ventilation systems, applying appropriate personal protective equipment (PPE), and risk management methods are the implementation cases to reduce exposure. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40201-024-00924-y.
Collapse
Affiliation(s)
- Safiye Ghobakhloo
- Department of Environmental Health Engineering, School of Health, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Khoshakhlagh
- Department of Occupational Health Engineering, School of Health, Kashan University of Medical Sciences, Kashan, Iran
| | - Gholam Reza Mostafaii
- Department of Environmental Health Engineering, School of Health, Kashan University of Medical Sciences, Kashan, Iran
| | | |
Collapse
|
92
|
Wang X, Liu J, Liu T, Fang C, Ding L, Li Q, Yang K, Wu X. Discovery of the pharmacodynamic material basis of Danggui Buxue Decoction in the treatment of diabetic kidney disease based on lipidomics regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156643. [PMID: 40215813 DOI: 10.1016/j.phymed.2025.156643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Danggui Buxue Decoction (DBD) is a formula used for treating diabetic kidney disease (DKD). However, the pharmacodynamic material basis of DBD in DKD therapy remains unclear, hindering its industrial development and innovation in drug formulations. PURPOSE Lipid metabolism disorder is a key pathological mechanism in DKD progression. This study employs lipidomics to elucidate and validate the pharmacodynamic material basis of DBD in treating DKD. METHODS Forty-eight male SD rats were used in the experiment, with 8 rats per group. The DKD model was constructed with a diet high in fat and sugar, together with intraperitoneal administration of low-dose STZ and unilateral nephrectomy. DBD was administered continuously for 10 weeks to assess its therapeutic efficacy on DKD. Lipid biomarkers in the DKD models were analyzed using lipidomics, while the transitional components in the blood of DBD-treated rats were characterized through UPLC-QE-Orbitrap MS. Potential pharmacodynamic substances were identified by correlating lipid biomarkers with active ingredients in vivo, followed by molecular docking and in vitro experiments to validate key pharmacodynamic components. RESULTS DBD significantly improved blood glucose, blood lipid levels, and renal function in DKD model rats. Lipidomics identified 37 lipid biomarkers in the DKD models, and DBD demonstrated a marked corrective effect on these biomarkers. In the therapeutically effective state, 91 blood transitional components of DBD were identified. Correlation analysis revealed 44 pharmacodynamic substances associated with DKD treatment, with ferulic acid, calycosin, astragaloside IV, and ligustilide being the key components. These substances acted by increasing the levels of SIRT1, PPARG, and ABCA1 proteins in lipid-deposited podocytes. CONCLUSION In conclusion, this study explained the scientific connotation of DBD treatment of DKD with modern scientific language from three aspects: pharmacodynamic evaluation, pharmacodynamic material basis and mechanism of action from the perspective of lipid metabolism balance for the first time, and provided an empirical basis for the modern application of traditional Chinese medicinal prescriptions.
Collapse
Affiliation(s)
- Xu Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Jing Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tingting Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Cheng Fang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Lin Ding
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; Qiqihar Medical University, Qiqihar 161006, China
| | - Qiyao Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Kaidi Yang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiuhong Wu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
93
|
Wang TH, Chou LF, Shen YW, Lin NC, Shih YH, Shieh TM. Mechanistic insights into temoporfin-based photodynamic therapy: Ferroptosis as a critical regulator under normoxic and hypoxic conditions in head and neck cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 267:113165. [PMID: 40267720 DOI: 10.1016/j.jphotobiol.2025.113165] [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: 03/04/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Temoporfin is a second-generation photosensitizer used in photodynamic therapy (PDT) for the clinical treatment of head and neck cancer. However, its role in inhibiting cancer cell viability under normoxic and hypoxic conditions remains unclear. The oral squamous cell carcinoma (OSCC) cell lines, SAS and OECM-1 were cultured under normoxic or hypoxic conditions to investigate temoporfin-based PDT-induced cell death and the underlying mechanisms. Cell viability was analyzed using the MTT assay. Intracellular reactive oxygen species (ROS) levels, cell apoptosis, intracellular ROS, iron levels, lipid peroxidation, and glutathione (GSH) levels were assessed by flow cytometry. The expression of proteins related to oxidative stress, apoptosis, autophagy, and ferroptosis was verified by western blotting. Results showed that increasing the temoporfin dose, absorption time, and illumination time was positively correlated with the inhibition of oral cancer cells. Hypoxic conditions attenuated the toxicity of temoporfin in cancer cells. OECM-1 cells were more sensitive to temoporfin than SAS cells. Temoporfin-based PDT-induced ROS exhibited similar trends to oxidative stress-inducing enzymes under both normoxic and hypoxic conditions and triggered cell autophagy and ferroptosis. Administration of the ferroptosis inhibitor BRD4770 under normoxic conditions reversed temoporfin-based PDT-induced reductions in glutathione peroxidase 4 (GPx4), increasing in light chain 3-II (LC3-II) and cleaved poly (ADP-ribose) polymerase (cleaved-PARP). This study confirms that hypoxia weakens the anticancer effects of temoporfin-based PDT and that ferroptosis plays a key role in temoporfin-based PDT-mediated cancer cell inhibition.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Biobank, Chang Gung Memorial Hospital at Linkou, No.5, Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; Graduate Institute of Health Industry Technology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No. 261, Wenhua 1st Rd., Guishan District, Taoyuan 33303, Taiwan; Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital at Linkou, No.5, Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; Graduate Institute of Natural Products, Chang Gung University, No.259, Wenhua 1st Rd., Guishan District, Taoyuan 33302, Taiwan.
| | - Li-Fang Chou
- Kidney Research Center, Chang Gung Memorial Hospital, Tao-Yuan 33305, Taiwan
| | - Yen-Wen Shen
- School of Dentistry, China Medical University, No. 100, Section 1, Jingmao Road, Beitun District, Taichung 406040, Taiwan.
| | - Nan-Chin Lin
- Department of Oral and Maxillofacial Surgery, Show Chwan Memorial Hospital, No.542, Sec 1 Chung-shan Rd., Changhua 500. Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, No.500, Lioufeng Rd., Wufeng, Taichung 413305, Taiwan.
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, No. 100, Section 1, Jingmao Road, Beitun District, Taichung 406040, Taiwan; Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou District, Taipei 112304, Taiwan.
| |
Collapse
|
94
|
Chakravarti B, Tomar MS, Qais FA, Raza S, Abdullah KM, Sharma G, Tewari A, Yadav A, Gupta P, Chattopadhyay N, Shrivastava A, Sinha RA, Siddiqui JA. Alpha lipoic acid modulates metabolic reprogramming in breast cancer stem cells enriched 3D spheroids by targeting phosphoinositide 3-kinase: In silico and in vitro insights. Biomed Pharmacother 2025; 187:118121. [PMID: 40327992 DOI: 10.1016/j.biopha.2025.118121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Breast cancer stem cells (BCSCs) are a unique subpopulation of tumor cells driving tumor resistance, progression, metastasis, and recurrence. Reprogrammed cellular metabolism and key signaling pathways, including Wnt/β-catenin, TGF-β, STAT3, and PI3K/AKT/mTOR pathway play a vital role in maintaining BCSCs. Importantly, PI3K/Akt/mTOR pathway regulates metabolism, survival, growth, and invasion, with PIK3CA, encoding the PI3K catalytic subunit p110α, the most frequently mutated gene in breast cancer. This study investigates the effects of alpha-lipoic acid (LA) on the metabolic profile of BCSCs, focusing on its interaction with PI3K signaling. LA was found to bind PI3K, disrupting cancer-associated metabolic pathways and significantly inhibiting BCSC metabolism. Metabolomic analysis of MCF-7 and MDA-MB-231-derived breast cancer spheroids showed LA-induced metabolic shifts. In MCF-7 spheroids, LA induced upaccumulation of 15 metabolites and downaccumulation of 5, while in MDA-MB-231 spheroids, it induced upaccumulation of 3 and downaccumulation of 16. LA also enhanced the sensitivity of breast cancer spheroids to doxorubicin (Dox), demonstrating a synergistic effect. Mechanistically, LA modulates the PI3K/Akt/mTOR pathway, impairing cell survival and proliferation. These findings highlight the potential of LA as a therapeutic agent for reprogramming cancer metabolism and enhancing chemotherapy efficacy. These results provide a strong rationale for incorporating LA into combination therapy strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Bandana Chakravarti
- Stem Cell/Cell Culture Lab, Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India.
| | - Manendra Singh Tomar
- Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Faizan Abul Qais
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Archana Tewari
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Abhishek Yadav
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Pratima Gupta
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Ashutosh Shrivastava
- Center for Advanced Research, Faculty of Medicine, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
95
|
Lu G, Tang Y, Chen O, Guo Y, Xiao M, Wang J, Liu Q, Li J, Gao T, Zhang X, Zhang J, Cheng Q, Kuang R, Gu J. Aberrant activation of p53-TRIB3 axis contributes to diabetic myocardial insulin resistance and sulforaphane protection. J Adv Res 2025; 72:467-484. [PMID: 39069209 DOI: 10.1016/j.jare.2024.07.025] [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: 07/02/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024] Open
Abstract
INTRODUCTION Insulin resistance (IR) is associated with multiple pathological features. Although p53- or TRIB3-orchestrated IR is extensively studied in adipose tissue and liver, the role of p53-TRIB3 axis in myocardial IR remains unknown, and more importantly target-directed therapies of myocardial IR are missing. OBJECTIVES Considering the beneficial effects of sulforaphane (SFN) on cardiovascular health, it is of particular interest to explore whether SFN protects against myocardial IR with a focus on the regulatory role of p53-TRIB3 axis. METHODS Mouse models including cardiac specific p53-overexpressing transgenic (p53-cTg) mice and Trib3 knockout (Trib3-KO) mice, combined with primary cardiomyocytes treated with p53 activator (nutlin-3a) and inhibitor (pifithrin-α, PFT-α), or transfected with p53-shRNA and Trib3-shRNA, followed by multiple molecular biological methodologies, were used to investigate the role of p53-TRIB3 axis in SFN actions on myocardial IR. RESULTS Here, we report that knockdown of p53 rescued cardiac insulin-stimulated AKT phosphorylation, while up-regulation of p53 by nutlin-3a or p53-cTg mice blunted insulin sensitivity in cardiomyocytes under diabetic conditions. Diabetic attenuation of AKT-mediated cardiac insulin signaling was markedly reversed by SFN in p53-Tgfl/fl mice, but not in p53-cTg mice. Importantly, we identified TRIB3 was elevated in p53-cTg diabetic mice, and confirmed the physical interaction between p53 and TRIB3. Trib3-KO diabetic mice displayed improved insulin sensitivity in the heart. More specifically, the AMPKα-triggered CHOP phosphorylation and degradation were essential for p53 on the transcriptional regulation of Trib3. CONCLUSION Overall, these results indicate that inhibiting the p53-TRIB3 pathway by SFN plays an unsuspected key role in the improvement of myocardial IR, which may be a promising strategy for attenuating diabetic cardiomyopathy (DCM) in diabetic patients.
Collapse
Affiliation(s)
- Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Ou Chen
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaohui Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jingjing Zhang
- Department of Cardiology at the First Hospital of China Medical University, and Department of Cardiology at the People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China
| | - Quanli Cheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Rong Kuang
- NMPA Key Laboratory for Animal Alternative Testing Technology of Cosmetics, Zhejiang Institute for Food and Drug Control, Hangzhou, Zhejiang 310004, China.
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
96
|
Ji P, Li Q, Zhang Y, Jin J, Zhang Y, Yuan Z, Shen G, Cao Q, Wu Y, Wang P, Liu W. The role of RAB12 in inhibiting osteogenic differentiation and driving metabolic dysregulation in osteoporosis. Life Sci 2025; 370:123590. [PMID: 40147529 DOI: 10.1016/j.lfs.2025.123590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
AIMS The osteogenic differentiation of mesenchymal stem cells (MSCs) is crucial in osteoporosis, and the metabolic level of the bone microenvironment directly affects metabolic dysregulation in postmenopausal women. RAB12 is a member of the small GTPase Rab family proteins, known to play an important role in autophagy. However, the role of RAB12 in the osteogenic differentiation of osteoporotic hMSCs remains unclear. MATERIALS AND METHOD Immunohistochemical staining was used to validate the high expression of RAB12 in aged osteoporotic mouse models and ovariectomized (OVX) mouse models. Co-immunoprecipitation (Co-IP) and LC-MS/MS were employed to explore downstream proteins that may interact with RAB12. Adenovirus containing RAB12 siRNA sequences was injected into the tail vein of OVX osteoporotic mice to analyze the impact of the RAB12/PCBP1/GLUT1 axis on MSC osteogenic differentiation. KEY FINDINGS We found that RAB12 expression is upregulated in elderly osteoporotic patients and in osteoporotic mouse models. RAB12 negatively regulates the osteogenic differentiation of hMSCs both in vivo and in vitro. RAB12 interacts with the PCBP1 protein, affecting its autophagic degradation when its expression levels change. RAB12 regulates the transcriptional level of GLUT1 by influencing the autophagic degradation of PCBP1, thereby affecting MSC's regulation of glucose uptake, which in turn impacts MSC osteogenic differentiation and metabolic changes. SIGNIFICANCE RAB12 negatively regulates osteogenic differentiation through the PCBP1/GLUT1 axis, affecting glucose metabolism levels in the bone microenvironment. RAB12 may serve as a potential target for the treatment of osteoporosis and postmenopausal metabolic dysregulation.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Quanfeng Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Yunhui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Jiahao Jin
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Yibin Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Zihao Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Guozhen Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Qian Cao
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China; Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China
| | - Yanfeng Wu
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China; Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China.
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China.
| |
Collapse
|
97
|
Abdel-Wahab BA, El-Shoura EAM, Habeeb MS, Aldabaan NA, Ahmed YH, Zaafar D. Piperazine ferulate impact on diabetes-induced testicular dysfunction: unveiling genetic insights, MAPK/ERK/JNK pathways, and TGF-β signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6719-6737. [PMID: 39671097 DOI: 10.1007/s00210-024-03654-y] [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: 04/06/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024]
Abstract
Diabetic testicular dysfunction (DTD) poses a significant threat to male reproductive health. This study delves into the potential of piperazine ferulate (PF), a natural phenolic compound, in alleviating DTD and sheds light on its underlying mechanisms in rats. Animals were divided into the control, PF, diabetic, and diabetic plus PF groups. Diabetes was induced in rats with a single intraperitoneal (i.p.) injection of streptozotocin (STZ) at 50 mg/kg. PF was administered at 50 mg/kg/day via i.p. injection for four weeks. Significant changes in sexual behavior were observed in diabetic rats, which additionally revealed lower serum levels of testosterone, FSH, and LH. The abnormalities in sperm count, viability, motility, and morphology occurred along with the demonstrated suppression of genes and protein expression related to spermatogenesis. Atrophy of the seminiferous tubules and extensive degeneration and necrosis of the germ and Leydig cells were highlighted by histopathological examination. The testicular function of diabetic rats was significantly improved after PF administration, evidenced by normalized testicular histology, increased testosterone levels, and enhanced sperm quality. In addition to reducing inflammatory cytokines, COX2, and NF-κB expression, pf administration elevated the antioxidant levels and Nrf2/HO-1 expression. Furthermore, key signaling pathways involved in testicular degeneration are regulated by PF. It promoted cell survival and tissue repair by activating the protective TGF-β signaling pathway and attenuating the MAPK/ERK/JNK signaling cascade, which in turn reduced inflammation and apoptosis. PF suppressed the expression of INSL3, SPHK1, CD62E, ANGPTL2, and miR-148a-5p, while increasing the expression of testicular genes like HSD17B1, DAZL, and S1P, addressing DTD. This study highlights the potential of PF to restore testicular function and fertility in diabetic males by modulating genetic and signaling pathways.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, P.O. Box 1988, Najran, Saudi Arabia.
| | - Ehab A M El-Shoura
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Mohammed S Habeeb
- Department of Pharmacology, College of Pharmacy, Najran University, P.O. Box 1988, Najran, Saudi Arabia
| | - Nayef A Aldabaan
- Department of Pharmacology, College of Pharmacy, Najran University, P.O. Box 1988, Najran, Saudi Arabia
| | - Yasmine H Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
98
|
Costa RM, Bruder-Nascimento A, Alves JV, Awata WMC, Singh S, Rodrigues D, Bruder-Nascimento T, Tostes RC. Beclin-1-dependent autophagy protects perivascular adipose tissue function from hyperaldosteronism effects. Am J Physiol Heart Circ Physiol 2025; 328:H1253-H1266. [PMID: 40327449 DOI: 10.1152/ajpheart.00829.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/13/2024] [Accepted: 04/22/2025] [Indexed: 05/08/2025]
Abstract
Hyperaldosteronism (HA), characterized by excessive production of aldosterone (Aldo), contributes to cardiovascular damage and perivascular adipose tissue (PVAT) dysfunction. Previous studies have shown that Aldo can impair autophagy in various tissues. However, it remains unclear whether this impairment occurs specifically in PVAT and whether it involves disruption of autophagic flux through Beclin-1 (BCN1), a key regulator of autophagosome formation and maturation. We hypothesize that BCN1-dependent autophagy plays a protective role in PVAT by limiting inflammation and preserving its anticontractile function in the context of HA. Male and female C57BL/6J [wild type (WT)] and BCN1 knock-in mice, aged 10-12 wk, underwent 14-day aldosterone infusion (600 µg/kg/day) using an osmotic minipump. Vascular function was assessed in PVAT-intact thoracic aortae, and blood pressure was monitored via radiotelemetry. HA disrupted PVAT autophagic flux, leading to the accumulation of LC3II/I and p62 proteins and reduced BCN1 expression/activity. In WT mice, PVAT exhibited an anticontractile effect, which was abolished by HA. In contrast, BCN1-knock-in mice were protected from this loss of PVAT function. HA also induced oxidative stress and inflammation in PVAT, as evidenced by increased reactive oxygen species generation and elevated mRNA levels of TNF-α, IL-6, IL-1β, and IL-17. These proinflammatory and prooxidative changes were not observed in BCN1-knock-in mice, indicating preserved PVAT homeostasis. Furthermore, pharmacological induction of autophagy via spermidine and activation of BCN1 with TB peptide improved PVAT function in HA-treated WT mice. Finally, BCN1-knock-in mice exhibited partial protection against HA-induced hypertension, highlighting the systemic vascular benefits of enhanced autophagic flux. In summary, our findings demonstrate that the activation of autophagy provides protection against HA-induced PVAT inflammation, dysfunction, and hypertension. Consequently, the activation of BCN1 could serve as a pharmacological strategy to prevent the harmful cardiovascular effects associated with HA.NEW & NOTEWORTHY Elevated aldosterone levels, as seen in primary hyperaldosteronism, obesity, and hypertension, impair autophagic flux in perivascular adipose tissue (PVAT), leading to increased inflammation and loss of anticontractile function. The Beclin-1-dependent autophagic pathway plays a key role in maintaining PVAT homeostasis and vascular tone. Disrupted autophagy contributes to oxidative stress and hypertension. Activating this pathway may offer a novel therapeutic strategy to mitigate aldosterone's harmful vascular effects in hypertension by restoring PVAT function and vascular inflammation.
Collapse
Affiliation(s)
- Rafael M Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Institute of Health Sciences, Federal University of Jatai, Jatai, Brazil
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
| | - Ariane Bruder-Nascimento
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
| | - Juliano V Alves
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Wanessa M C Awata
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Shubhnita Singh
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Daniel Rodrigues
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Bruder-Nascimento
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
99
|
Elariny HA, Atia HA, Abdallah MH, Khalifa AM, Abd Elmaaboud MA, Elkady MA, Kabel AM. Olmesartan attenuates doxorubicin-elicited testicular toxicity: The interaction between sirtuin-1, HMGB1/NLRP3 inflammasome/gasdermin D signaling, and AMPK/mTOR-driven autophagy. Life Sci 2025; 370:123545. [PMID: 40058574 DOI: 10.1016/j.lfs.2025.123545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND In the recent years, there has been an increased incidence of testicular toxicity associated with doxorubicin (DOX) use in cancer therapy. The mechanisms of this adverse effect may include induction of oxidative stress with augmentation of the inflammatory and the apoptotic signals in the testicular tissues. The ongoing research is directed towards the exploration of new agents that are capable of overcoming this health problem. This study was a trial to evaluate the efficacy of Olmesartan as a protective agent against DOX-induced testicular dysfunction in male rats. MATERIALS AND METHODS Forty adult male Sprague-Dawley rats were divided into control group, DOX-injected group, and three DOX-injected groups treated with olmesartan at 3 dose levels (1, 5, and 10 mg/kg/day). The effect of the different treatments was assessed at the biochemical and the morphological levels. KEY FINDINGS Olmesartan administered to DOX-treated rats induced dose-dependent restoration of the testicular weight and functions, normalization of the hormonal profile, augmentation of the antioxidant defenses, and potentiation of AMPK/mTOR-driven autophagy in comparison to rats treated with DOX alone. These effects were accompanied with a dose-dependent significant mitigation of the cellular events related to pyroptosis and inflammation and a significant amelioration of the testicular morphological changes induced by DOX. SIGNIFICANCE Olmesartan may represent a promising therapy for DOX-elicited testicular dysfunction, possibly via dose-dependent antioxidant, anti-pyroptotic, anti-inflammatory, and autophagy enhancing effects.
Collapse
Affiliation(s)
- Hemat A Elariny
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Hanan Abdelmawgoud Atia
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Marwa H Abdallah
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Amany M Khalifa
- Department of Pathology, College of Medicine, University of Ha'il, Ha'il 81442, Saudi Arabia
| | | | - Mennatallah A Elkady
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Ahmed M Kabel
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
100
|
Aleshcheva G, Salih S, Baumeier C, Escher F, Bock CT, Schultheiss H. Discovery of miRNAs unique to actively transcribed erythroparvovirus infection in heart failure patients. ESC Heart Fail 2025; 12:1872-1882. [PMID: 39970057 PMCID: PMC12055386 DOI: 10.1002/ehf2.15194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 02/21/2025] Open
Abstract
AIMS miRNAs, small non-coding RNAs, play key roles in gene regulation, cell differentiation and tissue development. They influence viral infection outcomes by directly interacting with viral genomes or modifying the host microenvironment. This study demonstrates miRNAs' ability to selectively suppress transcriptionally active erythroparvovirus, highlighting their potential in antiviral therapies. METHODS AND RESULTS Seventy-five endomyocardial biopsy (EMB) specimens from patients with unexplained heart failure were analysed. The samples included 19 with dilated cardiomyopathy and inflammation (DCMi), 12 with dilated cardiomyopathy (DCM), 25 with inflammation and active erythroparvovirus infection, 13 with active erythroparvovirus infection only and 6 from undiagnosed patients as controls. miRNA expression was measured using TaqMan assays. miR-98, miR-222, miR-106b and miR-197 were significantly upregulated in patients with transcriptionally active erythroparvovirus infection, independent of inflammation (P < 0.005). These miRNAs differentiated these patients from all other groups with over 90% specificity. CONCLUSIONS These specific miRNAs offer a novel diagnostic tool for active erythroparvovirus infections and hold promise as therapeutic targets, providing safer alternatives to traditional antiviral treatments.
Collapse
Affiliation(s)
- Ganna Aleshcheva
- Institute for Cardiac Diagnostics and Therapy (IKDT)BerlinGermany
| | - Sara Salih
- BHT – Berliner Hochschule für TechnikBerlinGermany
| | | | - Felicitas Escher
- Institute for Cardiac Diagnostics and Therapy (IKDT)BerlinGermany
- DHZC (German Heart Centre of Charité)BerlinGermany
- DZHK (German Centre for Cardiovascular Research), partner site BerlinBerlinGermany
| | - C. Thomas Bock
- Department of Infectious Diseases, Division of Viral Gastroenteritis and Hepatitis Pathogens and EnterovirusesRobert Koch InstituteBerlinGermany
| | | |
Collapse
|