201
|
Liu S, Yang X, Zheng S, Chen C, Qi L, Xu X, Zhang D. Research progress on the use of traditional Chinese medicine to treat diseases by regulating ferroptosis. Genes Dis 2025; 12:101451. [PMID: 40070365 PMCID: PMC11894312 DOI: 10.1016/j.gendis.2024.101451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/28/2024] [Accepted: 08/25/2024] [Indexed: 03/14/2025] Open
Abstract
Ferroptosis is an emerging form of programmed cell death triggered by iron-dependent lipid peroxidation. It is distinguished from other forms of cell death by its unique morphological changes and characteristic fine-tuned regulatory gene network. Since its pivotal involvement in the pathogenesis and therapeutic interventions of various diseases, such as malignant tumors, cardiovascular and cerebrovascular diseases, and traumatic disorders, has been well-established, ferroptosis has garnered significant attention in contemporary physiological and pathological research. For the advantage of alleviating the clinical symptoms and improving life quality, traditional Chinese medicine (TCM) holds a significant position in the treatment of these ailments. Moreover, increasing studies revealed that TCM compounds and monomers showed evident therapeutic efficacy by regulating ferroptosis via signaling pathways that tightly regulate redox reactions, iron ion homeostasis, lipid peroxidation, and glutathione metabolism. In this paper, we summarized the current knowledge of TCM compounds and monomers in regulating ferroptosis, aiming to provide a comprehensive review of disease management by TCM decoction, Chinese patent medicine, and natural products deriving from TCM through ferroptosis modulation. The formulation composition, chemical structure, and possible targets or mechanisms presented here offer valuable insights into the advancement of TCM exploration.
Collapse
Affiliation(s)
- Shuai Liu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Xianzhen Yang
- Urinary Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Sanxia Zheng
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Changjing Chen
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Lei Qi
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Xiangdong Xu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| | - Denglu Zhang
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
- Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China
| |
Collapse
|
202
|
Zhang Y, Tian R, Feng X, Xiao B, Yue Q, Wei J, Wang L. Overexpression of METTL3 in lung cancer cells inhibits radiation-induced bystander effect. Biochem Biophys Res Commun 2025; 761:151714. [PMID: 40184791 DOI: 10.1016/j.bbrc.2025.151714] [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: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Radiation-induced bystander effects (RIBE) increase the complexity of radiation therapy (RT). m6A modification is implicated in ionizing radiation damage. This study aims to investigate the RIBE and the mechanism after promoting m6A modification. METHODS Lung adenocarcinoma cells were treated to simulate a hypoxic and 0.5 Gy RT environment. The expression levels of METTL3, METTL14, and YTDHF2 were quantified by RT-qPCR. Paracellular clonogenicity and the expression of 53BP1 and γ-H2AX were assessed by immunofluorescence. The proliferative rate was evaluated by CCK-8. Probes were employed to measure ROS levels. Micronucleus formation was evaluated microscopically. m6A-mRNA/lncRNA microarrays, MERIP-PCR, RT-qPCR, and ELISA were utilized to assess m6A modification levels and the expression of inflammatory factors. RESULTS m6A modification levels were significantly diminished under hypoxic, low-dose irradiation conditions. The overexpression of METTL3 in irradiated cancer cells resulted in increased clonogenicity and proliferation of paracellular cells, suppressed the rate of micronucleus formation, and reduced DNA damage by modulating the inflammatory response. m6A-mRNA/lncRNA microarray analyses revealed a higher correlation of inflammatory molecules NF-κB and TRAF6. Further analysis demonstrated that the m6A modification levels of inflammation-related factors such as IL-6, TLR4, NF-κB2, and TRAF6 were significantly up-regulated, while their mRNA expression levels were notably decreased. Additionally, the expression of IL-10 and TGF-β was significantly reduced, with no significant changes observed in IL-1 expression. CONCLUSION The overexpression of METTL3 facilitated m6A modification and mitigated the inflammatory response, thereby promoting paracellular cloning and proliferation, inhibiting micronucleus formation, alleviating DNA damage, and achieving the objective of suppression of RIBE.
Collapse
Affiliation(s)
- Yong Zhang
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, China
| | - Rongrong Tian
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, China
| | - Xudong Feng
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, China
| | - Bin Xiao
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, China
| | - Qi Yue
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, China
| | - Jinling Wei
- The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Li Wang
- The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China.
| |
Collapse
|
203
|
Bensalem J, Teong XT, Hattersley KJ, Hein LK, Fourrier C, Dang LVP, Singh S, Liu K, Wittert GA, Hutchison AT, Heilbronn LK, Sargeant TJ. Intermittent time-restricted eating may increase autophagic flux in humans: an exploratory analysis. J Physiol 2025; 603:3019-3032. [PMID: 40345145 DOI: 10.1113/jp287938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Autophagy slows age-related pathologies and is stimulated by nutrient restriction in animal studies. However, this has never been shown in humans. We measured autophagy using a physiologically relevant measure of autophagic flux (flux of MAP1LC3B isoform II/LC3B-II in peripheral blood mononuclear cells in the context of whole blood) in 121 humans with obesity who were randomised to standard care (SC, control condition), calorie restriction (CR) or intermittent fasting plus time-restricted eating (iTRE) for 6 months. While the differences in change from baseline between groups was not significant at 2 months, we observed a significant difference in change from baseline between iTRE compared to SC at 6 months (P = 0.04, post hoc analysis). This effect may be driven partly by a tendency for autophagy to decrease in the SC group. The difference in change from baseline between CR and SC was not significant. Uncorrected analysis of correlations showed a negative relationship between change in autophagy and change in blood triglycerides. Data on the specificity and performance of the methods used to measure human autophagy are also presented. This shows autophagy may be increased by intermittent nutrient restriction in humans. If so, this is a demonstration that nutrient restriction can be used to improve a primary hallmark of biological ageing and provides a mechanism for how fasting could delay the onset of age-related disease. KEY POINTS: Autophagy slows biological ageing, and dysfunction of autophagy has been implicated in age-related disease - an effective way of increasing autophagy in cells and animal models is nutrient restriction. However, the impact of different types of nutrient restriction on physiological autophagic flux in humans has not been extensively researched. Here we measure the effect of intermittent time-restricted eating (iTRE) and calorie restriction on physiological autophagic flux in peripheral blood mononuclear cells. After 6 months, there was a significant difference in change from baseline between the iTRE and the standard care control group, with flux being higher in the iTRE group at this timepoint. However, there was no significant increase from baseline within the iTRE group, showing that although autophagy may be modified by nutrient restriction in humans, further studies are required.
Collapse
Affiliation(s)
- Julien Bensalem
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Xiao Tong Teong
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Kathryn J Hattersley
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Leanne K Hein
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Célia Fourrier
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Linh V P Dang
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Sanjna Singh
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Kai Liu
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Gary A Wittert
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Amy T Hutchison
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Leonie K Heilbronn
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Timothy J Sargeant
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| |
Collapse
|
204
|
Duan H, Siadat SH, Jasim SA, Bansal P, Kaur H, Qasim MT, Abosaoda MK, Aboqader Al-Aouadi RF, Suliman M, Ali Khiavi P. Therapeutic Potential of Exosomal miRNAs: New Insights and Future Directions. J Biochem Mol Toxicol 2025; 39:e70270. [PMID: 40272032 DOI: 10.1002/jbt.70270] [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: 12/24/2024] [Revised: 03/13/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Modern advancements in medicine include developing targeted drug delivery systems in the medical field, which are designed to unravel the potential of therapeutic products and overcome the barriers to the effectiveness of current approaches. Various nanopolymer carrier systems have been introduced in this regard, and the simple characteristics of extracellular vesicles have drawn special attention to their application as an effective drug delivery tool. Exosomes are very similar to transport vesicles and have a lipid-biomembrane covering an aqueous core. They also contain both hydrophilic and lipophilic substances and deliver their cargo to the desired targets. These properties enable exosomes to overcome some of the limitations of liposomes. Exosomes can easily diffuse into body fluids and remain in the bloodstream for a long time, crossing physiological barriers and entering cells. Exosomes, which contain a large volume of biomolecules, do not stimulate immune responses and do not accumulate in the liver or lungs instead of target tissues. Recent advancements in regenerative medicine have enabled scientists to utilize exosomes extracted from mesenchymal stem cells (MSCs), which possess significant regenerative abilities, for treating various diseases. The contents of these exosomes are crucial for both diagnosis and treatment, as they influence disease progression. Numerous in vitro studies have confirmed the safety, effectiveness, and therapeutic promise of exosomes in conditions such as cancer, neurodegenerative disorders, cardiovascular issues, and orthopedic ailments. This article explores the therapeutic potential of MSC-derived exosomes and outlines the essential procedures for their preparation.
Collapse
Affiliation(s)
- Haili Duan
- China Three Gorges University, Yichang City, China
| | | | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and medical technology, University of Al-maarif, Anbar, Iraq
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Maytham T Qasim
- Immunology and Physiology, College of Health and Medical Technology, Al-Ayen University, Iraq
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Al Diwaniyah, Iraq
| | | | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Payam Ali Khiavi
- Medicine Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
205
|
Zhang S, Zhang D, Xu K, Huang X, Chen Q, Chen M. The role of the farnesoid X receptor in diabetes and its complications. Mol Cell Biochem 2025; 480:2725-2736. [PMID: 39576464 DOI: 10.1007/s11010-024-05162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/10/2024] [Indexed: 01/06/2025]
Abstract
Diabetes is a metabolic disease in which tissues and organs are exposed to a hyperglycemic environment for a prolonged period. Long-term hyperglycemia can cause dysfunction of multiple organs and tissues in the body, leading to diabetic complications such as diabetic cardiomyopathy and diabetic nephropathy. Diabetes and its complications have become one of the key issues that seriously threaten the health of people worldwide. Farnesoid X receptor (FXR), as a metabolic regulator, has multiple functions in regulating insulin synthesis and secretion, insulin resistance, lipid metabolism, oxidative stress, inflammatory response, and fibrosis. It plays a key role in alleviating diabetes and its complications. In this review, we discuss the latest findings of FXR related to diabetes and its complications, focusing on its role in diabetes, diabetic nephropathy, diabetic cardiomyopathy, and diabetic liver injury. The aim is to better understand the role of FXR in diabetes and its complications and to provide new perspectives on the treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Shengnan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei, People's Republic of China
- School of Resources and Environmental Science and Engineering, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Dandan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei, People's Republic of China
| | - Kui Xu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei, People's Republic of China
- School of Clinical Medicine, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Xingqiong Huang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei, People's Republic of China
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei, People's Republic of China
| | - Mi Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, No. 88 Xianning Avenue, Xian'an District, Xianning, Hubei, People's Republic of China.
| |
Collapse
|
206
|
Zhang Y, Chen H, Chen Q, Zaitoun M, Cheng Y, Ge J, Feng Q. Acrolein-Triggered Ferroptosis and Protection by Intermittent Fasting via the AMPK/NRF2-CLOCK/BMAL1 Pathway. TOXICS 2025; 13:369. [PMID: 40423448 DOI: 10.3390/toxics13050369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025]
Abstract
Environmental pollution significantly exacerbates various diseases, particularly those affecting the cardiovascular and respiratory systems. Our previous studies have shown that acrolein, an environmental pollutant, promotes atherosclerosis by downregulating the circadian clock genes (CLOCK/BMAL1) and disrupting circadian rhythm. We have also found that intermittent fasting (IF), closely linked to the circadian clock, may mitigate atherosclerosis induced by acrolein. Ferroptosis, a newly identified form of regulated cell death, is associated with the acceleration of atherosclerotic development, but its relationship with the circadian clock is not well understood. In this study, we explored the potential of IF to alleviate ferroptosis by modulating the circadian clock. Our in vivo experiments revealed that IF reversed ferroptosis and upregulated CLOCK/BMAL1 in APOE-/- mice. In human umbilical vein endothelial cells (HUVECs), we discovered that acrolein-induced ferroptosis leads to cell death, while short-term starvation (STS, IF cell model) reversed this effect. Acrolein also suppressed the expression of AMP-activated protein kinase (AMPK), nuclear factor erythroid 2-related factor 2 (NRF2), and CLOCK/BMAL1, which were restored by subsequent STS treatments. Additionally, the overexpression of CLOCK/BMAL1 mitigated ferroptosis, consistent with findings from CLOCK gene knockout experiments. Notably, CLOCK/BMAL1 and AMPK/NRF2 were found to be mutually regulated. Concurrently, the AMPK and NRF2 signaling pathways may be interdependent and act in concert. In conclusion, our findings suggest that IF modulates the CLOCK/BMAL1-AMPK/NRF2 pathway to alleviate acrolein-induced ferroptosis, offering a potential strategy to address health issues related to environmental pollution.
Collapse
Affiliation(s)
- Yuandie Zhang
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Hong Chen
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Qianfeng Chen
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Margaret Zaitoun
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Ying Cheng
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Jierong Ge
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Qing Feng
- Department of Nutrition and Food Hygiene, Key Laboratory of Toxicology, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
207
|
Chu Y, Yang S, Chen X. Fibroblast growth factor receptor signaling in metabolic dysfunction-associated fatty liver disease: Pathogenesis and therapeutic targets. Pharmacol Ther 2025; 269:108844. [PMID: 40113178 DOI: 10.1016/j.pharmthera.2025.108844] [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: 01/08/2025] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as a significant hepatic manifestation of metabolic syndrome, with its prevalence increasing globally alongside the epidemics of obesity and diabetes. MAFLD represents a continuum of liver damage, spanning from uncomplicated steatosis to metabolic dysfunction-associated steatohepatitis (MASH). This condition can advance to more severe outcomes, including fibrosis and cirrhosis. Fibroblast growth factor receptors (FGFRs) are a family of four receptor tyrosine kinases (FGFR1-4) that interact with both paracrine and endocrine fibroblast growth factors (FGFs). This interaction activates the phosphorylation of tyrosine kinase residues, thereby triggering downstream signaling pathways, including RAS-MAPK, JAK-STAT, PI3K-AKT, and PLCγ. In the context of MAFLD, paracrine FGF-FGFR signaling is predominantly biased toward the development of liver fibrosis and carcinogenesis. In contrast, endocrine FGF-FGFR signaling is primarily biased toward regulating the metabolism of bile acids, carbohydrates, lipids, and phosphate, as well as maintaining the overall balance of energy metabolism in the body. The interplay between these biased signaling pathways significantly influences the progression of MAFLD. This review explores the critical functions of FGFR signaling in MAFLD from three perspectives: first, it examines the primary roles of FGFRs relative to their structure; second, it summarizes FGFR signaling in hepatic lipid metabolism, elucidating mechanisms underlying the occurrence and progression of MAFLD; finally, it highlights recent advancements in drug development aimed at targeting FGFR signaling for the treatment of MAFLD and its associated diseases.
Collapse
Affiliation(s)
- Yi Chu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Su Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
208
|
Sisto A, van Wermeskerken T, Pancher M, Gatto P, Asselbergh B, Assunção Carreira ÁS, De Winter V, Adami V, Provenzani A, Timmerman V. Autophagy induction by piplartine ameliorates axonal degeneration caused by mutant HSPB1 and HSPB8 in Charcot-Marie-Tooth type 2 neuropathies. Autophagy 2025; 21:1116-1143. [PMID: 39698979 PMCID: PMC12013449 DOI: 10.1080/15548627.2024.2439649] [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: 01/18/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
HSPB1 [heat shock protein family B (small) member 1] and HSPB8 are essential molecular chaperones for neuronal proteostasis, as they prevent protein aggregation. Mutant HSPB1 and HSPB8 primarily harm peripheral neurons, resulting in axonal Charcot-Marie-Tooth neuropathies (CMT2). Macroautophagy/autophagy is a shared mechanism by which HSPB1 and HSPB8 mutations cause neuronal dysfunction. Autophagosome formation is reduced in mutant HSPB1-induced pluripotent stem-cell-derived motor neurons from CMT type 2F patients. Likewise, the HSPB8K141N knockin mouse model, mimicking CMT type 2 L, exhibits axonal degeneration and muscle atrophy, with SQSTM1/p62-positive deposits. We show here that mouse embryonic fibroblasts isolated from a HSPB8K141N/green fluorescent protein (GFP)-LC3 model have diminished autophagosome production under conditions of MTOR inhibition. To correct the autophagic deficits in the HSPB1 and HSPB8 models, we screened by high-throughput autophagosome quantification the repurposing Spectrum Collection library for molecules that could boost the autophagic activity above the canonical MTOR inhibition. Hit compounds were validated on motor neurons obtained by differentiation of HSPB1P182L and HSPB8K141N patient-derived induced pluripotent stem cells, focusing on autophagy induction as well as neurite network density, axonal degeneration, and mitochondrial morphology. We identified molecules that specifically stimulate autophagosome formation in the HSPB8K141N cells, without affecting autophagy flux. Two top lead compounds induced autophagy and reduced axonal degeneration, thus promoting neuronal network maturation in the CMT2 patient-derived motor neurons. Based on these findings, the phenotypical screen revealed that piplartine rescued autophagy deficiencies in both the HSPB1 and HSPB8 models, demonstrating autophagy induction as an effective therapeutic strategy for CMT neuropathies and other chaperonopathies.
Collapse
Affiliation(s)
- Angela Sisto
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Tamira van Wermeskerken
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Pamela Gatto
- HTS Core Facility, University of Trento, Trento, Italy
| | - Bob Asselbergh
- Neuromics Support Facility, VIB - Center for Molecular Neurology, Antwerp, Belgium
- Neuromics Support Facility, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Vicky De Winter
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | | | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
209
|
Nàger M, Larsen KB, Bhujabal Z, Kalstad TB, Rössinger J, Myrmel T, Weinberger F, Birgisdottir AB. Mitophagy is induced in human engineered heart tissue after simulated ischemia and reperfusion. J Cell Sci 2025; 138:jcs263408. [PMID: 39912384 PMCID: PMC11959618 DOI: 10.1242/jcs.263408] [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: 06/26/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
The paradoxical exacerbation of cellular injury and death during reperfusion remains a problem in the treatment of myocardial infarction. Mitochondrial dysfunction plays a key role in the pathogenesis of myocardial ischemia and reperfusion injury. Dysfunctional mitochondria can be removed by mitophagy, culminating in their degradation within acidic lysosomes. Mitophagy is pivotal in maintaining cardiac homeostasis and emerges as a potential therapeutic target. Here, we employed beating human engineered heart tissue (EHT) to assess mitochondrial dysfunction and mitophagy during ischemia and reperfusion simulation. Our data indicate adverse ultrastructural changes in mitochondrial morphology and impairment of mitochondrial respiration. Furthermore, our pH-sensitive mitophagy reporter EHTs, generated by a CRISPR/Cas9 endogenous knock-in strategy, revealed induced mitophagy flux in EHTs after ischemia and reperfusion simulation. The induced flux required the activity of the protein kinase ULK1, a member of the core autophagy machinery. Our results demonstrate the applicability of the reporter EHTs for mitophagy assessment in a clinically relevant setting. Deciphering mitophagy in the human heart will facilitate development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mireia Nàger
- Division of Cardiothoracic and Respiratory Medicine, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Kenneth B. Larsen
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
- Department of Medical Biology, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Zambarlal Bhujabal
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Trine B. Kalstad
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Judith Rössinger
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Truls Myrmel
- Division of Cardiothoracic and Respiratory Medicine, University Hospital of North Norway, 9019 Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Asa B. Birgisdottir
- Division of Cardiothoracic and Respiratory Medicine, University Hospital of North Norway, 9019 Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| |
Collapse
|
210
|
Guerra G, Venturelli E, Polymeropoulos A, Segrado F, Morelli D, Rinaldi S, Agnoli C, Sieri S. Comprehensive assessment of matrix effects in metallomics: Evaluating 27 metals in serum, heparine-plasma-, EDTA-plasma and citrate-plasma by ICP-MS analysis. J Trace Elem Med Biol 2025; 89:127667. [PMID: 40339194 DOI: 10.1016/j.jtemb.2025.127667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/27/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Metallomics analysis is essential for studying environmental metal exposure and distinguishing between healthy and diseased individuals in large-scale studies. Inductively coupled plasma mass spectrometry (ICP-MS) is widely used due to its high sensitivity and ability to analyze multiple elements across a broad concentration range. AIM Developed to address key challenges in large-scale epidemiological studies, this method specifically focuses on evaluating the performance of metals measurement in biobank blood matrices that may not be optimized for metallomics, providing key insights for biobank planning METHODS: An ICP-MS-based analytical method using a "dilute and shoot" approach was developed to target 27 elements. The study assessed the behavior of these elements in serum and different plasma types (EDTA, citrate, and heparin) based on limits of detection (LOD), limits of quantification (LOQ), precision, accuracy, and quality controls. RESULTS Heparin plasma and serum provided the most consistent measurements, with most elements exhibiting a coefficient of variation below 15 %. Citrated and EDTA plasma displayed higher variability, likely due to contamination from collection tubes and metal-anticoagulant interactions. Among 13 certified elements, Mg, K, Fe, Cu, Se, Co, and Ni matched reference values, while Ca, Zn, Cr, Mn, and Al were lower, and Hg was higher. Of the 14 elements with indicative values, all except Cd, which was significantly lower, aligned with Seronorm™ references. CONCLUSION These findings highlight the impact of plasma matrices on metal measurements in biobank studies and emphasize the importance of selecting appropriate blood matrices and collection tubes to ensure accurate and reliable elemental analysis in large-scale epidemiological research.
Collapse
Affiliation(s)
- Giulia Guerra
- s.c. Epidemiologia e Prevenzione, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Elisabetta Venturelli
- s.s.d. Ricerche Nutrizionali e Metabolomica, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Alessio Polymeropoulos
- s.s. Biostatistica per la Ricerca Clinica, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Francesco Segrado
- s.s.d. Ricerche Nutrizionali e Metabolomica, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Daniele Morelli
- s.c. Medicina di laboratorio, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Sabina Rinaldi
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France.
| | - Claudia Agnoli
- s.c. Epidemiologia e Prevenzione, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Sabina Sieri
- s.c. Epidemiologia e Prevenzione, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
211
|
Xie S, Yang H. SP1 activates AKT3 to facilitate the development of diabetic nephropathy. J Endocrinol Invest 2025; 48:1269-1281. [PMID: 39786707 DOI: 10.1007/s40618-025-02530-7] [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: 09/18/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe complication of diabetes mellitus and has the complex pathogenesis. The previous study reported that protein kinase Bγ (AKT3) was involved in DN progression. Our aim was to explore the detailed mechanisms of AKT3 in DN development. METHODS RT-qPCR was performed to measure the levels of specificity protein 1 (SP1) and AKT3. Mesangial cells were treated with high glucose (30 mM) to form DN cell model in vitro. Western blot was conducted to detect the protein expression of AKT3, SP1, fibrosis-related proteins, and AKT/mTOR pathway-related proteins. Cell proliferation and inflammation were evaluated via MTT, EdU staining, and ELISA assays, respectively. Oxidative stress was determined via measuring ROS and MDA levels. ChIP and dual-luciferase reporter assays were carried out to verify the relationship between SP1 and AKT3. C57BL/6 mice-treated with streptozotocin for 5 days were used to establish DN mouse model in vivo, and HE and Masson staining were conducted to evaluate pathological changes of mouse kidney tissues. RESULTS AKT3 and SP1 were highly expressed in DN kidney tissues and HG-induced mesangial cells. AKT3 depletion could relieve HG treatment-caused cell damage of mesangial cells through repressing cell proliferation, fibrosis, inflammation and oxidative stress. SP1 can bind to the promoter of AKT3 and serve as a translation regulation factor of AKT3. SP1 overexpression worsened HG treatment-caused cell damage of mesangial cells. Moreover, AKT3 upregulation could block the suppressive effects of SP1 depletion on cell proliferation, fibrosis, inflammation and oxidative stress in HG-induced mesangial cells. SP1 depletion reduced AKT3 expression to inactivate the AKT/mTOR pathway in HG-induced mesangial cells. Besides, AKT3 knockdown inhibited the activation of the AKT/mTOR pathway to hamper the development of DN in mice through alleviating fibrosis and inflammation in vivo. CONCLUSION Our results indicated that SP1 activated AKT3 and AKT/mTOR pathway to promote mesangial cell proliferation, fibrosis, inflammation and oxidative stress, thereby facilitating DN development.
Collapse
Affiliation(s)
- Shanshan Xie
- Department of Endocrinology, Nanshi Hospital of Nanyang, No. 130, West Zhongzhou Road, Nanyang, 473065, China
| | - Han Yang
- Department of Endocrinology, Nanshi Hospital of Nanyang, No. 130, West Zhongzhou Road, Nanyang, 473065, China.
| |
Collapse
|
212
|
Yan Z, Han J, Mi Z, Wang Z, Fu Y, Wang C, Dang N, Liu H, Zhang F. GPNMB disrupts SNARE complex assembly to maintain bacterial proliferation within macrophages. Cell Mol Immunol 2025; 22:512-526. [PMID: 40038549 PMCID: PMC12041529 DOI: 10.1038/s41423-025-01272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Xenophagy plays a crucial role in restraining the growth of intracellular bacteria in macrophages. However, the machinery governing autophagosome‒lysosome fusion during bacterial infection remains incompletely understood. Here, we utilize leprosy, an ideal model for exploring the interactions between host defense mechanisms and bacterial infection. We highlight the glycoprotein nonmetastatic melanoma protein B (GPNMB), which is highly expressed in macrophages from lepromatous leprosy (L-Lep) patients and interferes with xenophagy during bacterial infection. Upon infection, GPNMB interacts with autophagosomal-localized STX17, leading to a reduced N-glycosylation level at N296 of GPNMB. This modification promotes the degradation of SNAP29, thus preventing the assembly of the STX17-SNAP29-VAMP8 SNARE complex. Consequently, the fusion of autophagosomes with lysosomes is disrupted, resulting in inhibited cellular autophagic flux. In addition to Mycobacterium leprae, GPNMB deficiency impairs the proliferation of various intracellular bacteria in human macrophages, suggesting a universal role of GPNMB in intracellular bacterial infection. Furthermore, compared with their counterparts, Gpnmbfl/fl Lyz2-Cre mice presented decreased Mycobacterium marinum amplification. Overall, our study reveals a previously unrecognized role of GPNMB in host antibacterial defense and provides insights into its regulatory mechanism in SNARE complex assembly.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Jinghong Han
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zihao Mi
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenzhen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yixuan Fu
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Chuan Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ningning Dang
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Furen Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
213
|
Damiani I, Solberg EH, Iyer M, Cheng P, Weldy CS, Kim JB. Environmental pollutants and atherosclerosis: Epigenetic mechanisms linking genetic risk and disease. Atherosclerosis 2025; 404:119131. [PMID: 39986958 PMCID: PMC12034486 DOI: 10.1016/j.atherosclerosis.2025.119131] [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: 08/28/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Over the past half-century, significant strides have been made to identify key risk factors, genetic mechanisms, and treatments for atherosclerosis. Yet, coronary artery disease (CAD) remains a leading global public health challenge. While the heritability of CAD is well-documented, there is increasing focus on the role of environmental exposures, such as smoking, air pollution, and heavy metals, on global CAD risk. Recent research has shed light on the interplay between genetic variation and environmental factors, offering insights into gene-environment (GxE) interactions. Moreover, emerging evidence suggests that environmental toxicants can profoundly impact the epigenome, altering gene regulation beyond the genetic sequence itself, revealing novel mechanisms underlying disease. Epigenetic changes - such as modifications in DNA methylation, chromatin structure, and non-coding RNA function - are now recognized as key molecular determinants of atherosclerosis. These observations have created a foundational paradigm that environment, genetics, and epigenetic mechanisms influence risk through a highly complex interaction regulating cellular phenotype, pathology, and disease progression. In this review, we explore the mechanisms by which environmental exposures influence the epigenome and contribute to the regulation of atherosclerotic disease. Additionally, we examine the transgenerational epigenetic effects of these exposures on disease risk. Advancing our understanding of these mechanisms is essential for informing public health strategies aimed at mitigating harmful environmental exposures and reducing the global burden of cardiovascular disease.
Collapse
Affiliation(s)
- Isabella Damiani
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Elena Hurtado Solberg
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Meghana Iyer
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chad S Weldy
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA; Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
214
|
Shang W, Geng X, Sun X, Fan X, Li A, Zhang C, Kang Y, Liang Y, Zhang J. Non-coding RNAs modulate pyroptosis in diabetic cardiomyopathy: A comprehensive review. Int J Biol Macromol 2025; 309:142865. [PMID: 40188918 DOI: 10.1016/j.ijbiomac.2025.142865] [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: 01/01/2025] [Revised: 02/07/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of heart failure (HF) among individuals with diabetes, presenting a significant medical challenge due to its complex pathophysiology and the lack of targeted therapies. Pyroptosis, a pro-inflammatory form of programmed cell death (PCD), is the predominant mode of cell death in the primary resident cells involved in DCM. It has been reported to be critical in DCM's onset, progression, and pathogenesis. Non-coding RNAs (ncRNAs), diverse transcripts lacking protein-coding potential, are essential for cellular physiology and the progression of various diseases. Increasing evidence indicates that ncRNAs are pivotal in the pathogenesis of DCM by regulating pyroptosis. This observation suggests that targeting the regulation of pyroptosis by ncRNAs may offer a novel therapeutic approach for DCM. However, a comprehensive review of this topic is currently lacking. Our objective is to elucidate the regulatory role of ncRNAs in pyroptosis associated with DCM and to elucidate the relationships among these factors. Additionally, we explored how ncRNAs influence pyroptosis and contribute to the pathophysiology of DCM. By doing so, we aim to identify new research targets for the clinical diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Wenyu Shang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Xiaofei Geng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Xitong Sun
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Xinbiao Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Aolin Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Chi Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Yuxin Kang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Yongchun Liang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
| | - Junping Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China.
| |
Collapse
|
215
|
Xie Y, Liang B, Meng Z, Guo R, Liu C, Yuan Y, Mu W, Wang Y, Cao J. Downregulation of HSPB1 and MGST1 Promotes Ferroptosis and Impacts Immune Infiltration in Diabetic Cardiomyopathy. Cardiovasc Toxicol 2025; 25:719-734. [PMID: 40053272 DOI: 10.1007/s12012-025-09982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/26/2025] [Indexed: 03/12/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Current therapies do not adequately resolve this problem and focus only on the optimal level of blood glucose for patients. Ferroptosis plays an important role in diabetes mellitus and cardiovascular diseases. However, the role of ferroptosis in DCM remains unclear. Differentially expressed ferroptosis-related genes (DE-FRGs) were identified by intersection of the GSE26887 dataset and the Ferroptosis Database. The associations between the DE-FRGs and immune cells in DCM, estimated via the CIBERSORTx algorithm, were analysed. Flow cytometry (FCM) was used to evaluate the infiltration of immune cells in myocardial tissues. The expression of DE-FRGs, glutathione peroxidase 4 and solute carrier family 7 member 11 was examined via real-time quantitative PCR and Western blotting. Three DE-FRGs were identified: heat shock protein family B (small) member 1 (HSPB1), microsomal glutathione S-transferase 1 (MGST1) and solute carrier family 40 member 1 (SLC40A1), which are closely linked to immune cells in DCM. In vivo, the levels of CD8 + T cells, B cells and regulatory T (Treg) cells were significantly decreased in the DCM group, whereas the levels of CD4 + T cells, M1 cells, M2 cells and monocytes were increased. Diabetes significantly decreased HSPB1 and MGST1 levels and increased ferroptosis compared with the Normal group. Furthermore, the ferroptosis inhibitor ferrostatin-1 (Fer-1) alleviated high-fat diet (HFD)-induced cardiomyocyte injury and rescued ferroptosis. These findings suggest that the ferroptosis-related genes HSPB1 and MGST1 are closely related to immune cell infiltration and may be therapeutic targets for DCM.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Animals
- Diabetic Cardiomyopathies/immunology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/metabolism
- Male
- Down-Regulation
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Disease Models, Animal
- Molecular Chaperones/genetics
- Molecular Chaperones/metabolism
- Humans
- Signal Transduction
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- HSP27 Heat-Shock Proteins/genetics
- HSP27 Heat-Shock Proteins/metabolism
- Mice, Inbred C57BL
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/metabolism
- Databases, Genetic
Collapse
Affiliation(s)
- Yaoli Xie
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Bin Liang
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Zhijun Meng
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Rui Guo
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Caihong Liu
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yi Yuan
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Wei Mu
- Department of Interventional Therapy for Tumor and Vascular Interventional Therapy, The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Yajing Wang
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Jimin Cao
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
216
|
Waligórski MPR, Janiak MK. Comment on "The scientific nature of the linear no-threshold (LNT) model used in the system of radiological protection" by A. Wojcik and F. Zölzer. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2025; 64:329-331. [PMID: 39982465 DOI: 10.1007/s00411-025-01112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025]
Affiliation(s)
- Michael P R Waligórski
- Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland, and National Institute of Oncology, Krakow Division, Krakow, Poland
| | - Marek K Janiak
- Professor emeritus, former head of Dept of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., Warszawa, 01-163, Poland.
| |
Collapse
|
217
|
Abavisani M, Tafti P, Khoshroo N, Ebadpour N, Khoshrou A, Kesharwani P, Sahebkar A. The heart of the matter: How gut microbiota-targeted interventions influence cardiovascular diseases. Pathol Res Pract 2025; 269:155931. [PMID: 40174272 DOI: 10.1016/j.prp.2025.155931] [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: 09/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
The human body is habitat to a wide spectrum of microbial populations known as microbiota, which play an important role in overall health. The considerable research has mostly focused on the gut microbiota due to its potential to impact numerous physiological functions and its correlation with a variety of disorders, such as cardiovascular diseases (CVDs). Imbalances in the gut microbiota, known as dysbiosis, have been linked to the development and progression of CVDs through various processes, including the generation of metabolites like trimethylamine-N-oxide and short-chain fatty acids. Studies have also looked at the idea of using therapeutic interventions, like changing your diet, taking probiotics or prebiotics, or even fecal microbiota transplantation (FMT), to change the gut microbiota's make-up and how it works in order to prevent or treat CVDs. Exploring the cause-and-effect connection between the gut microbiota and CVDs offers a hopeful path for creating innovative microbiome-centered strategies to prevent and cure CVDs. This review presents an in-depth review of the correlation between the gut microbiota and CVDs, as well as potential therapeutic approaches for manipulating the gut microbiota to enhance cardiovascular health.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pourya Tafti
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khoshroo
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pardesh, India; University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
218
|
Kurmangaliyeva S, Baktikulova K, Tkachenko V, Seitkhanova B, Shapambayev N, Rakhimzhanova F, Almagambetova A, Kurmangaliyev K. An Overview of Hexavalent Chromium-Induced Necroptosis, Pyroptosis, and Ferroptosis. Biol Trace Elem Res 2025; 203:2619-2635. [PMID: 39287767 DOI: 10.1007/s12011-024-04376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Heavy metals are common environmental industrial pollutants. Due to anthropogenic activity, chromium, especially its hexavalent form [Cr(VI)], is a widespread environmental contaminant that poses a threat to human health. In this review paper, we summarize the currently reported molecular mechanisms involved in chromium toxicity with a focus on the induction of pro-inflammatory non-apoptotic cell death pathways such as necroptosis, pyroptosis, and ferroptosis. The review highlights the ability of chromium to induce necroptosis, pyroptosis, and ferroptosis revealing the signaling pathways involved. Cr(VI) can induce RIPK1/RIPK3-dependent necroptosis both in vitro and in vivo. Chromium toxicity is associated with pyroptotic NLRP3 inflammasome/caspase-1/gasdermin D-dependent secretion of IL-1β and IL-18. Furthermore, this review emphasizes the role of redox imbalance and intracellular iron accumulation in Cr(VI)-induced ferroptosis. Of note, the crosstalk between the investigated lethal subroutines in chromium-induced toxicity is primarily mediated by reactive oxygen species (ROS), which are suggested to act as a rheostat determining the cell death pathway in cells exposed to chromium. The current study provides novel insights into the pro-inflammatory effects of chromium, since necroptosis, pyroptosis, and ferroptosis affect inflammation owing to their immunogenic properties linked primarily with damage-associated molecular patterns. Inhibition of these non-apoptotic lethal subroutines can be considered a therapeutic strategy to reduce the toxicity of heavy metals, including chromium.
Collapse
Affiliation(s)
- Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kristina Baktikulova
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan.
| | - Viktoriya Tkachenko
- State Institution "Republican Scientific and Practical Centre of Sports, " 8 Narochanskaya St, Minsk, Republic of Belarus
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Al-Farabi Sq, Shymkent, Republic of Kazakhstan
| | - Nasriddin Shapambayev
- Department of General Practitioner - 1, Khoja Akhmet Yasawi International Kazakh-Turkish University, 7/7 Baitursynov St, Shymkent, Republic of Kazakhstan
| | - Farida Rakhimzhanova
- Department of Microbiology, NCJSC "Semey Medical University, " 103 Abay St, Semey, Republic of Kazakhstan
| | - Altyn Almagambetova
- Department of Phthisiology and Dermatovenerology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kairat Kurmangaliyev
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| |
Collapse
|
219
|
Saez Lancellotti TE, Avena MV, Funes AK, Bernal-López MR, Gómez-Huelgas R, Fornes MW. Exploring the impact of lipid stress on sperm cytoskeleton: insights and prospects. Nat Rev Urol 2025; 22:294-312. [PMID: 39528754 DOI: 10.1038/s41585-024-00952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The decline in male fertility correlates with the global rise in obesity and dyslipidaemia, representing significant public health challenges. High-fat diets induce metabolic alterations, including hypercholesterolaemia, hepatic steatosis and atherosclerosis, with detrimental effects on testicular function. Testicular tissue, critically dependent on lipids for steroidogenesis, is particularly vulnerable to these metabolic disruptions. Excessive lipid accumulation within the testes, including cholesterol, triglycerides and specific fatty acids, disrupts essential sperm production processes such as membrane formation, maturation, energy metabolism and cell signalling. This leads to apoptosis, impaired spermatogenesis, and abnormal sperm morphology and function, ultimately compromising male fertility. During spermiogenesis, round spermatids undergo extensive reorganization, including the formation of the acrosome, manchette and specialized filamentous structures, which are essential for defining the final sperm cell shape. In this Perspective, we examine the impact of high-fat diets on the cytoskeleton of spermatogenic cells and its consequences to identify the mechanisms underlying male infertility associated with dyslipidaemia. Understanding these processes may facilitate the development of therapeutic strategies, such as dietary interventions or natural product supplementation, that aim to address infertility in men with obesity and hypercholesterolaemia. The investigation of cytoskeleton response to lipid stress extends beyond male reproduction, offering insights with broader implications.
Collapse
Affiliation(s)
- Tania E Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina.
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina.
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain.
| | - María V Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Abi K Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu), Instituto de Histología y Embriología (IHEM), Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María-Rosa Bernal-López
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Gómez-Huelgas
- Servicio de Medicina Interna, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel W Fornes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM), Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
220
|
Zhang J, Cheng L, Jiang S, Zhu D. Machine learning based identification of anoikis related gene classification patterns and immunoinfiltration characteristics in diabetic nephropathy. Sci Rep 2025; 15:15271. [PMID: 40312440 PMCID: PMC12046048 DOI: 10.1038/s41598-025-99395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 04/21/2025] [Indexed: 05/03/2025] Open
Abstract
Anoikis and immune cell infiltration are pivotal factors in the pathophysiological mechanism of diabetic nephropathy (DN), yet a comprehensive understanding of the mechanism is lacking. This work aimed to pinpoint distinctive anoikis-related genes (ARGs) in DN and delve into their impact on the immune landscape. Three datasets (GSE30528, GSE47184, and GSE96804) were downloaded from the gene expression omnibus (GEO) dataset. Differentially expressed genes (DEGs) were identified using the "limma" package, while ARGs were obtained from GSEA, GeneCard, and Harmonizome datasets. The intersection of DEGs and ARGs was analyzed for Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. The CIBERSORT algorithm was employed to estimate the infiltration percentage of 22 immune cell types in DN renal tissue. Subsequently, the least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF) algorithms were adopted to screen key ARGs related to DN. After that, receiver operating characteristic (ROC) analysis was employed to assess the diagnostic accuracy of each gene and the real-time quantitative polymerase chain reaction (RT-qPCR) was adopted to quantitatively detect the expression of biomarkers in DN cell models. Finally, correlations between key genes and immune cell infiltration were analyzed, and a competitive endogenous ribonucleic acid (RNA) (ceRNA) network based on key genes was constructed. A total of 59 DEARGs were identified. GO functional annotation enrichment analysis revealed their involvement in kidney development, extracellular matrix (ECM), cytoplasmic vesicle cavity, immunoinflammatory response, and cytokine effect. KEGG pathway analysis indicated that MAPK, PI3K -Akt, IL -17, TNF, and HIF- 1 signaling pathways are critical for DN. In addition, seven key genes, including PDK4, S100A8, HTRA1, CHI3L1, WT1, CDKN1B, and EGF, were screened by machine learning algorithm. Most of these genes exhibited low expression in renal tissue of DN patients and positive correlation with neutrophils, and their expressions were verified in an external dataset cell model. The ceRNA analysis suggested potential regulatory pathways (H19/miR-15b-5p/PDK4 and KCNQ1T1/miR-1207-3p/WT1) influencing early DN progression. This work provided a comprehensive analysis of the role of DEARGs in DN for the first time, offering valuable insights for further understanding the disease mechanism and guiding clinical diagnosis, treatment, and research of DN.
Collapse
Affiliation(s)
- Jing Zhang
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China
| | - Lulu Cheng
- Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Shan Jiang
- Traditional Chinese Medicine Department of Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Duosheng Zhu
- The Third Department of Jiaozhou City Traditional Chinese Medicine Hospital, Jiaozhou, 266300, Shandong, China.
| |
Collapse
|
221
|
Chen Z, Zhong M, Lin Y, Zhang W, Zhu Y, Chen L, Huang Z, Luo K, Lu Z, Huang Z, Yan Y. METTL7B-induced histone lactylation prevents heart failure by ameliorating cardiac remodelling. J Mol Cell Cardiol 2025; 202:64-80. [PMID: 40068772 DOI: 10.1016/j.yjmcc.2025.03.006] [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: 09/07/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
INTRODUCTION Lactylation is important for a variety of biological activities. It is reported that Class I histone deacetylases (HDAC1-3) are histone lysine delactylases. However, the role of lactylation in cardiac remodelling remains uncertain. OBJECTIVES To explore a novel regulator of lactylation and elucidate their functional mechanisms in cardiac remodelling and heart failure. METHODS GSE36961, GSE141910 and GSE174691 related to HCM (hypertrophic cardiomyopathy) were separately acquired from Gene expression Omnibus. Candidate genes related to both HCM and histone lactylation were determined by the intersection of DEGs (differentially expressed genes) and module genes sifted by WGCNA (Weighted Gene Co-Expression Network Analysis). METTL7B was screened out and its expression in hypertrophic myocardium was measured by qRT-PCR and western blotting. Furthermore, immunofluorescence, immunoprecipitation, and RNA pull-down assays were utilized to identify the biological functions of METTL7B. The myocardial biopsy of HCM and transverse aortic constriction (TAC) mouse model were performed to analyze the effects of METTL7B on cardiac remodelling in vivo. RESULTS We observed that the expression of METTL7B was down-regulated in hypertrophic myocardium, and the lactylation level was increased during the early stage and falling rapidly in the process of cardiac remodelling. Furthermore, we demonstrated that sodium lactate (NALA) administration fulfil a protective role on cardiac remodelling, and METTL7B alleviates cardiac remodelling and improves heart function by maintaining the activation of histone lactylation possibly at the later stage. Impressively, METTL7B suppressed the expression of USP38 via m6A dependent mRNA degradation, resulting in increasing ubiquitylation of HDAC3, which is a proven histone lysine delactylases. CONCLUSION We identifed METTL7B as a potential therapeutic target for myocardial remodelling and showed that it played a critical role in the promotion of myocardial lactylation, which is beneficial for improvement of cardiac function and attenuation of cardiac remodelling.
Collapse
Affiliation(s)
- Ziqi Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Meijun Zhong
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yuhui Lin
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Wei Zhang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yinghong Zhu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Lin Chen
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Ziyao Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Kaiyuan Luo
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Zhifeng Lu
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Zhaoqi Huang
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Yi Yan
- Department of Cardiology, Translational Research Center for Regenerative Medicine and 3D Printing Technologies, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
222
|
Zeng J, Yuan L, Chen G, Qi Y, Qie X, Jin Y, Chen Y, Li H. The ferroptosis of sertoli cells inducing blood-testis barrier damage is produced by oxidative stress in cryptorchidism. Free Radic Biol Med 2025; 232:97-106. [PMID: 40032029 DOI: 10.1016/j.freeradbiomed.2025.02.043] [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: 12/24/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Oxidative Stress (OS) is the main cause of damage to the Blood-Testis Barrier (BTB) in cryptorchidism, which seriously endangers male reproductive health. It is well known that the OS induced ferroptosis is an important cause of dysfunction in the body. However, it is still unknown whether BTB damage in cryptorchidism leads to ferroptosis of Sertoli cells. We establishing the cryptorchidism model through surgery to avoid the complex effects of drugs on the model animals, combined with in vitro culture of the primary Sertoli cells for validation, and the methods of immunofluorescence staining, Western blotting and Prussian blue staining were used to study the oxidative stress in cryptorchidism. The effects of ferroptosis of Sertoli cells inducing BTB damage caused by OS in cryptorchidism were analyzed. We found that the inhibition of Nrf-2/keap-1/HO-1 pathway resulted in decreased expression levels of Glutathione Peroxidase 4 (GPX4), Ferroportin 1 (FPN1), and increased expression of Ferritin light chain (FTL) protein. Our research further confirms that inhibiting ferroptosis reduced BTB damage by reflecting a decrease expression of Zonula Occludens protein 1 (ZO-1), Occludin and Claudin-11 protein caused by OS. In addition, we found that the testosterone (T) secretion disorders and the supplementation of T can alleviate the damage of the BTB in cryptorchidism, and this effect is achieved through the Androgen Receptor (AR). In conclusion, our study found that the inhibition of Nrf-2/keap-1/HO-1 pathway in testis and the reduction of Tight junction proteins (TJs) ZO-1, Occludin and Claudin-11 protein expression levels in cryptorchidic mice, indicated that the cryptorchidism triggering a serious reproductive disorder, and one of the important reasons is the OS induced ferroptosis of Sertoli cells, which ultimately leads to the damage of the BTB. This findings may have important implications in the field of male reproductive disorders.
Collapse
Affiliation(s)
- Jianlin Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Ligang Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China.
| | - Guojuan Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Huangzhong District Animal Disease Prevention and Control Center, Xining, 811600, China
| | - Yumei Qi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Xiaolong Qie
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Yajuan Jin
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Yulu Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Haijun Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| |
Collapse
|
223
|
Masoumvand M, Ramezani E, Eshaghi Milasi Y, Baradaran Rahimi V, Askari VR. New horizons for promising influences of sulforaphane in the management of metabolic syndrome: a mechanistic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4933-4946. [PMID: 39702599 DOI: 10.1007/s00210-024-03706-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
The disorder known as metabolic syndrome (MetS) represents a substantial threat to society since it is linked to a higher risk of heart disease, diabetes, stroke, and other health issues. Although there is no known cure for metabolic syndrome, lifestyle changes in diet and physical activity can help. Sulforaphane (SFN), a compound in cruciferous vegetables, has been recognized as a promising treatment for addressing metabolic syndrome. The information was compiled after a thorough search of four databases, PubMed, Scopus, Web of Sciences, and Google Scholar. This analysis includes 86 studies that include clinical and nonclinical SFN investigations in diseases connected to metabolic syndrome. Research has shown that sulforaphane is a prospective treatment option for obesity, type 2 diabetes mellitus (T2-DM), and associated metabolic disorders due to its capacity to regulate fatty acid production and glucose management. Many molecular processes have been investigated, including activating nuclear factor erythroid 2-related factor 2(Nrf2), activating nuclear factor erythroid 2(NF-E2), reducing reactive oxygen species, and upregulating insulin receptor substrate 1(IRS-1) and other suggested mechanisms. The current review established many facts in favor of SFN's prospective benefits in metabolic syndrome. More studies in this field involving human studies are necessary to determine whether SFN may effectively treat metabolic syndrome.
Collapse
Affiliation(s)
- Mohammad Masoumvand
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Ramezani
- Department of Nutrition, Faculty of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
224
|
Zhang Z, Hu K, Fang Z, Wang S, Chen J, Yin D, Zhang C, Ma G. Acacetin reduces endoplasmic reticulum stress through the P-eNOS/PERK signaling pathway to attenuate MGO-induced vascular endothelial cell dysfunction. FEBS Open Bio 2025; 15:793-809. [PMID: 39927486 PMCID: PMC12051029 DOI: 10.1002/2211-5463.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Diabetic macrovascular disease is one of the most morbid and deadly complications of diabetes. Endothelial dysfunction plays a key role in diabetic macrovascular complications and endothelial cell apoptosis is one of the key indicators of endothelial dysfunction. Methylglyoxal (MGO), a highly reactive dicarbonyl compound generated during glycolysis, is related to the pathogenesis of cardiovascular diseases and may also promote endothelial dysfunction. Acacetin (ACA) is a naturally occurring flavonoid that can inhibit apoptosis, oxidative stress and inflammation to slow the progression of coronary heart disease; however, its effects on endothelial dysfunction are unknown. The present study investigated whether ACA may ameliorate MGO-induced endothelial dysfunction in human umbilical vein endothelial cells. The results revealed that the viability and apoptosis of human umbilical vein endothelial cells induced by MGO decreased after ACA treatment, which was reflected in the expression levels of the apoptosis-related proteins b-cell lymphoma 2 (Bcl-2)-associated death, Bcl-2-associated x protein and Bcl-2. Additionally, ACA downregulated the expression of key protein markers of MGO-induced endoplasmic reticulum stress, physical evidence recovery kit, eukaryotic initiation factor 2 alpha, activating transcription factor 4 and C/EBP homologous protein, with which calcium inward currents may be closely related. ACA significantly downregulated the MGO-induced expression of the cytosolic calcium channel proteins stromal interaction molecule 1, transient receptor potential canonical 1, ORAI calcium release-activated calcium modulator 1, transient receptor potential vanilloid 1 and 4, and the trans-endoplasmic reticulum membrane protein, transmembrane and coiled-coil domains 1. Finally, ACA increased the expression of phosphorylated endothelial nitric oxide synthase (Ser1177), thus increasing the expression of nitric oxide in endothelial cells. Overall, acacetin could reduce endoplasmic reticulum stress through the phosphorylated-endothelial nitric oxide/physical evidence recovery kit signaling pathway to attenuate MGO-induced vascular endothelial cell dysfunction. These findings may hold potential for the use of acacetin in diabetic macrovascular complications.
Collapse
Affiliation(s)
- Zhen Zhang
- School of PharmacyAnhui University of Chinese MedicineHefeiAnhuiChina
| | - Kaien Hu
- School of PharmacyAnhui University of Chinese MedicineHefeiAnhuiChina
| | - Zhaohui Fang
- Department of EndocrineThe First Hospital Affiliated to Anhui University of Chinese MedicineHefeiAnhuiChina
| | - Sihai Wang
- Department of EndocrineThe First Hospital Affiliated to Anhui University of Chinese MedicineHefeiAnhuiChina
| | - Jie Chen
- School of PharmacyAnhui University of Chinese MedicineHefeiAnhuiChina
| | - Dengke Yin
- School of PharmacyAnhui University of Chinese MedicineHefeiAnhuiChina
| | - Caiyun Zhang
- School of PharmacyAnhui University of Chinese MedicineHefeiAnhuiChina
| | - Gefei Ma
- School of PharmacyAnhui University of Chinese MedicineHefeiAnhuiChina
- Anhui Qimen Institute of SnakebiteHuangshanChina
| |
Collapse
|
225
|
Deng J, Qin J, Song G, Li C, Tang W, Tang Y, Xiao X, Wu L, He S, Zhou Y, Li J, Wang Y. The potential of low-intensity pulsed ultrasound to apply the long-term ovary protection from injury induced by 4-vinylcyclohexene diepoxide through inhibiting granulosa cell apoptosis. Bioeng Transl Med 2025; 10:e10744. [PMID: 40385545 PMCID: PMC12079353 DOI: 10.1002/btm2.10744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 12/07/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025] Open
Abstract
The potential of low-intensity pulsed ultrasound (LIPUS) in regulating ovarian function has been demonstrated; however, there is a lack of scientific evidence regarding the long-term efficacy of LIPUS in treating ovarian injury and understanding its regulatory mechanisms. In this study, 4-vinylcyclohexene diepoxide (VCD) was used to induce ovarian injury in rats, and LIPUS was applied to target the damaged ovarian tissues. The research aimed to investigate the long-term protective effect of LIPUS against ovum toxicity induced by VCD and elucidate the associated molecular mechanisms. During the experiment, HE staining was employed for observing the morphology and structure of the ovary, while protein sequencing was utilized for identifying and confirming the molecular mechanism through which LIPUS restores the damaged ovarian structure. The long-term effectiveness of LIPUS in protecting against ovarian injury was evaluated through ELISA, estrous cycle monitoring, fertility testing, and behavioral analysis. The results indicated that LIPUS effectively restored the structure of damaged ovaries. Both in vivo and in vitro studies revealed that this protective effect may be attributed to LIPUS inhibiting apoptosis of ovarian granulosa cells (GCs) by regulating Daxx-mediated ASK1/JNK signaling pathway. Subsequent functional tests demonstrated significant improvements in sex hormone secretion and regulation of estrous cycle within 6 cycles following LIPUS treatment. Additionally, there was a notable increase in offspring numbers after mating. Behavioral analysis revealed that LIPUS effectively alleviated menopausal symptoms resulting from ovarian injury including mood fluctuations, cognitive behavior changes, and reduced muscle excitability levels. These findings suggest that beneficial effects of LIPUS may help reduce VCD-induced ovarian damage with long-term efficacy.
Collapse
Affiliation(s)
- Juan Deng
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Juan Qin
- Department of Obstetrics and Gynecology, Guiyang Maternal and Child Health Care HospitalGuizhou Medical UniversityGuizhouChina
| | - Guolin Song
- Department of EmergencyThe Second Affiliated Hospital of Guizhou University of Traditional Chinese MedicineGuizhouChina
| | - Chenghai Li
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Wentao Tang
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Yilin Tang
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Xinfang Xiao
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Liu Wu
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Sicheng He
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Yiqing Zhou
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Junfen Li
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Yan Wang
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| |
Collapse
|
226
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [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: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
227
|
Zhao J, Han M, Nie Q, Wen X, Geng H, Zou Y, Li S, Xie W. Network pharmacology combined with experimental analysis to explore the mechanism of the XinShuaiNing formula on heart failure. 3 Biotech 2025; 15:110. [PMID: 40191450 PMCID: PMC11965065 DOI: 10.1007/s13205-025-04288-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/22/2025] [Indexed: 04/09/2025] Open
Abstract
This study was conducted to elucidate the mechanism of action of the Traditional Chinese Medicine XinShuaiNing (XSN) formula in CHF based on network pharmacology. A total of 489 compounds in the XSN formula were screened. These compounds predicted 778 targets. A search of CHF yielded 789 corresponding targets, and 151 intersections between the potential targets of the XSN formula and CHF, involving AKT1, AGT, eNOS, and VEGF. Abdominal aortic coarctation (AAC) was used to establish a CHF rat model, and isoproterenol-induced H9c2 cells to establish a myocardial injury cell model. The results showed that the XSN formula downregulated ET-1, BNP, and Hcy and upregulated the ALB levels and also relieved cardiac histopathological damage. The XSN formula reduced the content of pro-inflammatory factors and inhibited the apoptosis of cardiomyocytes. In addition, the expression of fibronectin, α-SMA, collagen 1, and collagen 3 was downregulated by XSN formula treatment, and the fibrotic areas of myocardial tissue were reduced. The XSN formula promoted phosphorylation of AKT1-induced VEGF and eNOS signaling and inhibited AGT signaling. Besides, the XSN formula can affect the apoptosis of H9c2 cells by affecting AKT1, AGT, eNOS, and VEGF. The XSN formula regulates inflammatory factors by inducing phosphorylation of AKT1, upregulating eNOS and VEGF, and downregulating AGT to protect cardiomyocytes from apoptosis and myocardial fibrosis to alleviate CHF. In conclusion, this study identified the target of XSN prescription through network pharmacology screening and experimental validation and confirmed its anti-inflammatory, antiapoptotic, and antifibrotic effects.
Collapse
Affiliation(s)
- Jue Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingjun Han
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Nie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Wen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyu Geng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Zou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Songyun Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
228
|
Mainkar G, Ghiringhelli M, Zangi L. The Potential of RNA Therapeutics in Treating Cardiovascular Disease. Drugs 2025; 85:659-676. [PMID: 40175855 DOI: 10.1007/s40265-025-02173-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Despite significant advances in cardiology over the past few decades, cardiovascular diseases (CVDs) remain the leading cause of global mortality and morbidity. This underscores the need for novel therapeutic interventions that go beyond symptom management to address the underlying causal mechanisms of CVDs. RNA-based therapeutics represent a new class of drugs capable of regulating specific genetic and molecular pathways, positioning them as strong candidates for targeting the root causes of a wide range of diseases. Moreover, owing to the vast diversity in RNA form and function, these molecules can be utilized to induce changes at different levels of gene expression regulation, making them suitable for a broad array of medical applications, even within a single disease context. Several RNA-based therapies are currently being investigated for their potential to address various CVD pathologies. These include treatments aimed at promoting cardiac revascularization and regeneration, preventing cardiomyocyte apoptosis, reducing harmful circulating cholesterols and fats, lowering blood pressure, reversing cardiac fibrosis and remodeling, and correcting the genetic basis of inherited CVDs. In this review, we discuss the current landscape of RNA therapeutics for CVDs, with an emphasis on their classifications, modes of action, advancements in delivery strategies and considerations for their implementation, as well as CVD targets with proven therapeutic potential.
Collapse
Affiliation(s)
- Gayatri Mainkar
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Matteo Ghiringhelli
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lior Zangi
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
229
|
Abolhasani S, Ahmadi Y, Fattahi D, Rostami Y, Chollou KM. microRNA-Mediated Regulation of Oxidative Stress in Cardiovascular Diseases. J Clin Lab Anal 2025; 39:e70017. [PMID: 40183484 PMCID: PMC12078765 DOI: 10.1002/jcla.70017] [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: 12/10/2024] [Revised: 02/08/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are the leading cause of mortality globally, often linked to oxidative stress. MicroRNAs (miRNAs) have emerged as significant regulators of oxidative stress within the cardiovascular system. OBJECTIVE This review examines the complex relationship between miRNAs and oxidative stress, clarifying their effects on gene expression pathways related to ROS production and detoxification in CVDs. METHODS From August to October 2024, we conducted a comprehensive search of PubMed, Scopus, Web of Science, and Google Scholar for studies published between 2014 and 2024 investigating the role of miRNAs in oxidative stress and cardiovascular diseases. RESULTS Specific miRNAs have been identified as critical regulators in the pathophysiology of CVDs, with distinct expression patterns correlated with conditions such as hypertension, coronary artery disease, and heart failure. For instance, miR-21 exacerbates oxidative stress by targeting genes essential for redox homeostasis, while miR-210 promotes endothelial cell survival under hypoxic conditions by mitigating ROS levels. CONCLUSION The reciprocal relationship between miRNAs and oxidative stress highlights the potential for therapeutic interventions targeting miRNA expression and activity in managing CVDs. Understanding these molecular mechanisms is vital for developing innovative strategies to address oxidative damage in cardiac tissues and improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Sakhavat Abolhasani
- Department of Basic Sciences and HealthSarab Faculty of Medical SciencesSarabEast AzerbaijanIran
| | - Yasin Ahmadi
- Department of Medical Laboratory ScienceKomar University of Science and TechnologySulaymaniyahKurdistan RegionIraq
| | - Davood Fattahi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityLiverpoolUK
| | - Yavar Rostami
- Department of Basic Sciences and HealthSarab Faculty of Medical SciencesSarabEast AzerbaijanIran
| | - Khalil Maleki Chollou
- Department of Basic Sciences and HealthSarab Faculty of Medical SciencesSarabEast AzerbaijanIran
| |
Collapse
|
230
|
Zhang L, Wang R, Xue Q, Wang Y, Xu J, Wang C, Fang X, Gao S, Zhang H, Guo L. Bioinformatic Analysis for Exploring Target Genes and Molecular Mechanisms of Cadmium-Induced Nonalcoholic Fatty Liver Disease and Targeted Drug Prediction. J Appl Toxicol 2025; 45:858-865. [PMID: 39806544 DOI: 10.1002/jat.4752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/19/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
Cadmium (Cd) is a widely available metal that has been found to have a role in causing nonalcoholic fatty liver disease (NAFLD). However, the detailed toxicological targets and mechanisms by which Cd causes NAFLD are unknown. Therefore, the present work aims to reveal the main targets of action, cellular processes, and molecular pathways by which cadmium causes NAFLD. As shown in the bioinformatics analysis, there were 74 main targets of action for cadmium-induced NAFLD, hemopoietic cell kinase (HCK), EPH receptor A2 (EPHA2), MYC proto-oncogene (MYC), lysyl oxidase (LOX), dipeptidyl peptidase 7 (DPP7), nuclear factor erythroid 2-related factor 2 (NFE2L2), dual specificity phosphatase 6 (DUSP6), CD2 cytoplasmic tail binding protein 2 (CD2BP2), notch receptor 3 (NOTCH3), and phospholipase A2 group IVA (PLA2G4A) were screened as core genes. Testing these core genes in other databases, three differentially expressed genes, HCK, MYC, and DUSP6 were verified and used as targets for drug prediction in DsigDB; decitabine and retinoic acid were screened as potential therapeutic drugs for NAFLD based on the p-value and the combined score. The results of molecular docking showed that the predicted drugs can bind well to the core targets. In conclusion, cadmium is associated with NAFLD; the identified cadmium-toxicity targets, HCK, MYC, and DUSP6, may serve as biomarkers for the diagnosis of NAFLD and predicted drugs, decitabine and retinoic acid may have a potential role in the treatment of NAFLD.
Collapse
Affiliation(s)
- Le Zhang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Rui Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Qian Xue
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Yongjie Wang
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao, China
| | - Jiayunzhu Xu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Chaofan Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Xin Fang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Shidi Gao
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| | - Haiying Zhang
- Department of Pathology, College of Basic Medical Sciences, the Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Li Guo
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
231
|
Acuna N, Park SY, Conti DV, Stern MC, Wu AH, Cheng I, Wilkens LR, Shu XO, Setiawan VW. Circulating microRNAs and alcohol consumption in the multiethnic cohort study. Alcohol 2025; 124:105-110. [PMID: 39880058 PMCID: PMC11977456 DOI: 10.1016/j.alcohol.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 12/20/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Excessive alcohol consumption is a significant public health concern and contributes to liver diseases and cancer. Modifiable lifestyle factors including alcohol consumption can influence circulating microRNAs (miRNAs), which are increasingly used as biomarkers for early disease detection. Yet limited studies have identified miRNAs associated with alcohol intake, particularly in multiethnic populations. We aimed to assess the association of alcohol consumption and circulating miRNAs in the Multiethnic Cohort Study. Participants (N = 917) had alcohol consumption data collected at baseline and miRNA data collected at follow-up. Negative binomial models were used to assess the association between alcohol consumption (continuous and categorical [nondrinkers: 0 g of ethanol/day; light drinkers: <28 g of ethanol/day for men and <14 g of ethanol/day for women; and heavy drinkers: ≥28 g of ethanol/day for men and ≥14 g of ethanol/day for women]) and miRNAs. Stratified analyses also examined categories by sex, race/ethnicity, smoking status, and body mass index. Overall, there were 52% non-drinkers, 37 % light drinkers, and 11 % were heavy drinkers. We did not detect an association of miRNAs with alcohol intake in continuous models after correcting for multiple comparisons. However, we did find an inverse association for light drinkers [incidence rate ratio (IRR) = 0.59, p = 8.21E-04] and heavy drinkers (IRR = 0.44, p = 1.47E-03) compared to nondrinkers for miR-451a. Additionally, miR-320e (IRR = 0.63, p = 1.61E-03) had an inverse association with alcohol intake for light drinkers compared to nondrinkers. Subgroup analysis also suggested there were differences by subgroups, underscoring that miRNAs used to detect chronic diseases may be subgroup specific. When stratified by case-control status, we found that among controls both light and heavy drinkers were associated with miR-451a. We identified an association for light and heavy drinkers with miR-451a and mir-320e, miRNAs associated with cancers and liver diseases, in comparison to nondrinkers.
Collapse
Affiliation(s)
- Nicholas Acuna
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Song-Yi Park
- Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mariana C Stern
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Lynne R Wilkens
- Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Veronica Wendy Setiawan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
232
|
Zhu Y, Kang D, Bai X, Luo P, Du B, Li B. Plasma Zinc Levels in Patients with Diabetic Nephropathy: Is there a Relationship with NLRP3 Inflammasome Activation and Renal Prognosis? Biol Trace Elem Res 2025; 203:2550-2560. [PMID: 39264567 DOI: 10.1007/s12011-024-04369-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Zinc is an essential trace element, and impaired zinc homeostasis may be associated with inflammation in patients with diabetic nephropathy (DN). We investigated the influence of zinc level on nod-like receptor nucleotide-binding domain and leucine-rich repeat pyrin-3 domain (NLRP3) inflammasome expression and renal prognosis in patients with DN. We recruited 90 patients definitively diagnosed with DN by renal biopsy and 40 healthy controls. Zinc, NLRP3, interleukin (IL)-1β, and IL-18 levels were detected in blood samples, and the correlations between these parameters were assessed. Receiver operating characteristic (ROC) curve and decision curve analysis (DCA) evaluated the predictive value of zinc and the NLRP3 inflammasome for DN. Furthermore, patients with DN were divided into low- and normal-zinc groups to observe differences in clinical indicators and identify expression of inflammatory-related factors in renal tissue. Kaplan-Meier survival curves predicted the impact of zinc levels on renal prognosis. We found that the plasma zinc concentration in patients with DN was lower, while NLRP3, IL-1β, and IL-18 levels were higher than were those in patients without DN (P < 0.05). Zinc level was negatively correlated with NLRP3, IL-1β, and IL-18 levels (P < 0.01). Zinc and the NLRP3 inflammasome were predictive of DN, but their combination improved the diagnostic value. The DCA curve demonstrated a good positive net benefit in the combined model. Compared to patients with low zinc levels, patients with normal zinc levels had lower expression of NLRP3 inflammasome and a better prognosis. Zinc has a protective effect on DN and may affect NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Dedong Kang
- Department of Anatomy, Showa University School of Medicine, Shinagawa-Ku, Tokyo, 142-8555, Japan
| | - Xiaoying Bai
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Bing Du
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China.
| |
Collapse
|
233
|
Yan R, Tan HW, Zhao XY, Wu JY, Zhong QH, Wang XY, Cai NL, Xu YM, Lau ATY. A methionine/aspartate-rich synthetic peptide delineated from N-terminal region of nucleophosmin protein effectively protects against cadmium-induced toxicity. ENVIRONMENT INTERNATIONAL 2025; 199:109443. [PMID: 40273557 DOI: 10.1016/j.envint.2025.109443] [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: 08/28/2024] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025]
Abstract
Cadmium (Cd) is a widespread toxic heavy metal, and exposure to Cd is a growing environmental health concern. The molecular mechanism of Cd cytotoxicity is complicated and still not well understood, and treatment options for Cd cytotoxicity are lacking. Currently, only a limited number of Cd-targeted proteins have been identified. Here, we used Cd-immobilized metal ion affinity chromatography (Cd-IMAC) coupled with LC-MS/MS technique to detect putative Cd-binding proteins in human cells, and nucleophosmin (NPM1) was identified as the top Cd-binding protein. We found that Cd bound exclusively to the methionine/aspartate (M/D)-rich region (MEDSMDMDM) of NPM1, and NPM1 was essential for Cd-induced apoptosis. Furthermore, Cd could trigger intracellular nucleolar stress by causing nucleoplasmic translocation of NPM1 and decreasing pre-rRNA levels through binding to the M/D-rich region of NPM1. Interestingly, we discovered that a short peptide containing only the M/D-rich region of NPM1 could effectively mitigate Cd toxicity, both in vitro and in vivo. Specifically, the synthetic M/D-rich peptide demonstrated significant protection against Cd toxicity, particularly in the liver. It significantly reduced Cd concentration, suppressed the upregulation of blood ALT and AST levels, and alleviated liver inflammation in Cd-exposed BALB/c mice. This study reveals a novel mechanism of Cd cytotoxicity through NPM1-regulated nucleolar stress and apoptosis. Additionally, it identifies a short peptide with strong clinical potential to counteract Cd toxicity.
Collapse
Affiliation(s)
- Rui Yan
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Heng Wee Tan
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Xiao-Yun Zhao
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Jia-Yi Wu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Qiu-Hua Zhong
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Xiu-Yun Wang
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Na-Li Cai
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Yan-Ming Xu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China.
| | - Andy T Y Lau
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China.
| |
Collapse
|
234
|
Tuerxun G, Li W, Abudurexiti G, Zhuo Q, Tuerdi A, Abulizi G. EPB41L3 Inhibits the Progression of Cervical Cancer Via the ERK/p38 MAPK Signaling Pathway. Mol Biotechnol 2025; 67:1958-1967. [PMID: 38907071 DOI: 10.1007/s12033-024-01172-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/02/2024] [Indexed: 06/23/2024]
Abstract
This study was aimed to uncover the character and potential regulatory mechanism of EPB41L3 in cervical cancer (CC). CC cells were injected into BALB/c nude mice (female) to construct a xenograft tumor model. Real-time quantitative polymerase chain reaction (qRT-PCR) and western blot were performed to evaluate the expression of EPB41L3, ERK/p38 MAPK signal markers in CC tissues and cells. Cell counting kit-8 (CCK-8) and Transwell was applied to analyze the viability, invasion, and migration of CC cell lines. EPB41L3 was substantially decreased both in CC tissues and cells. Cell viability, invasion, and migration of CC cells were reduced by overexpressing EPB41L3. Bioinformatics analysis prerdicted that EPB41L3 was strongly related to the ERK/p38 MAPK pathway. Compared with Ad-nc mice, the volume and weight of tumors and ERK/p38 MAPK signal markers were down-regulated in Ad-EPB41L3 mice. After knocking down EPB41L3 with EPB41L3 siRNA (siEPB41L3), the ERK/p38 MAPK pathway was activated. Moreover, SB203580 treatment reversed the effect of EPB41L3 silencing on the improvement in viability, migration, and invasion of CC cells. EPB41L3 suppresses the progression of CC via activating the ERK/p38 MAPK pathway. EPB41L3 may serve as an effective therapeutic target for CC.
Collapse
Affiliation(s)
- Gulixian Tuerxun
- Fifth Department of Gynecologic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, No. 789 East Suzhou Street, Xinshi District, Urumqi, 830000, China
| | - Wenyun Li
- Fifth Department of Gynecologic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, No. 789 East Suzhou Street, Xinshi District, Urumqi, 830000, China
| | - Guligeina Abudurexiti
- Fifth Department of Gynecologic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, No. 789 East Suzhou Street, Xinshi District, Urumqi, 830000, China
| | - Qian Zhuo
- Fifth Department of Gynecologic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, No. 789 East Suzhou Street, Xinshi District, Urumqi, 830000, China
| | - Awahan Tuerdi
- Maternal and Child Health Service Center Shayibake District, Urumqi, China
| | - Guzalinuer Abulizi
- Fifth Department of Gynecologic Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, No. 789 East Suzhou Street, Xinshi District, Urumqi, 830000, China.
| |
Collapse
|
235
|
Luo JQ, Wang L, Liao ZQ, Lu BX, Luo CY, He HY, Ou Yang ZH, Duan SB, He SH, Wei AY, Zhang HB. Adipose stem cells ameliorate erectile dysfunction in diabetes mellitus rats by attenuating ferroptosis through NRP1 with SLC7A11 interaction. Free Radic Biol Med 2025; 232:40-55. [PMID: 40020883 DOI: 10.1016/j.freeradbiomed.2025.02.041] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Adipose stem cells (ADSCs) have garneVred increasing attention for their potential to treat diabetes mellitus erectile dysfunction (DMED), but the underlying molecular mechanisms remain unclear. The aim of this study was to identify and investigate the key cytokines and mechanisms by which ADSCs improve erectile function in DMED rats. METHODS We performed in vivo and in vitro assays, including rat erectile function assessment, cell co-culture, cytokine microarray screening and co-immunoprecipitation to investigate the role of ADSCs in improving erectile function in DMED rats. RESULTS Our analyses confirmed the occurrence of ferroptosis in the corpus cavernosum of DMED rats, while ADSCs treatment significantly restored erectile function and improved relevant indicators of ferroptosis. In vitro assays further indicated that corpus cavernosum smooth muscle cells (CCSMCs) co-cultured with ADSCs exhibited enhanced resistance to ferroptosis, with notably lower levels of cytoplasmic and lipid reactive oxygen species compared to the ferroptosis inducer Erastin-treated group. Mechanistic studies revealed that Neuropilin 1 (NRP1) may be a key molecule in ADSCs to improve erectile function in DMED rats. Furthermore, NRP1 in CCSMCs can interact with solute carrier family 7 member 11 (SLC7A11) to enhance the function of the glutamate-cysteine countertransport (Xc-) system and ferroptosis resistance in CCSMCs. CONCLUSION In conclusion, our findings indicate that NRP1 is a key molecule for ADSCs treatment to alleviate ferroptosis and improve erectile function in DMED rats, providing a promising target for DMED treatment and prognosis.
Collapse
Affiliation(s)
- Jun-Qi Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Li Wang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zi-Qi Liao
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Bing-Xin Lu
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Cai-Yu Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Hai-Yang He
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Zhi-Han Ou Yang
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Song-Bo Duan
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Shu-Hua He
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China.
| | - An-Yang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China; Department of Urology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, Guangdong, 511340, China.
| | - Hai-Bo Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
236
|
Joshi A, Mandal R. Review Article on Molecular Basis of Zinc and Copper Interactions in Cancer Physiology. Biol Trace Elem Res 2025; 203:2458-2470. [PMID: 39215955 DOI: 10.1007/s12011-024-04356-5] [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: 06/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Various clinical manifestations associated with measurable abnormalities of Zn and Cu in serum and tissue were determined in Cancer-Patients (CP), and therefore, these two metals are drawing more and more attention presently than ever before. Cancer is a disease of uncontrolled-abnormal-cell-division with invasion-potential which was exhibited to occur due to dys-regulation/dys-homeostasis of fundamental-biological-pathways (FBP) including antioxidant-enzyme-defense-system, anti-inflammatory and immune-systems, and DNA-damage-repair-system in the human-body resulting in generation of chronic-oxidative-stress induced DNA-damage and gene-mutations, inflammation and compromised immune-system, tumor-induced increased angiogenesis, and inhibition of apoptosis processes. Zn and Cu were recognized to be the most crucial components of FBP and imbalance in Zn/Cu ratios in CP asserted to generate chronic toxicity in human body through various mechanisms including increased chronic oxidative stress linked compromised DNA integrity and gene mutations due to malfunctioning of DNA damage repair enzymes; increased angiogenesis process due to Zn- and Cu-binding proteins metallothionein and ceruloplasmin-induced enhanced expression of tumor growth factors; and elevation in inflammatory response which was further shown to down/upregulate gene expression of multiple Zn transporter proteins leading to dys-homeostasis of intracellular Zn concentrations, and it was determined to disturb the equilibrium between cell growth and division, proliferation, differentiation, and apoptosis processes which lead to cancer progression. Moreover, Zn was reported to affect matrix metalloproteinase activity and influence immune system cells to respond differently to different cytokines and enhance immune-suppressive effects accelerating the angiogenesis, invasion, and metastasis potential in cancer. Further, the most significant use of serum Cu/Zn ratio was recommended in clinical diagnosis, prognosis, tumor stage, patient survival, and cancer follow-up studies which need further investigations to elucidate and explore their roles in cancer physiology for clinical perspective.
Collapse
Affiliation(s)
- Amit Joshi
- PG Department of Biotechnology and Microbial Biotechnology, Sri Guru Gobind Singh College, Chandigarh, UT, India
| | - Reshu Mandal
- PG Department of Zoology, Sri Guru Gobind Singh College, Chandigarh, UT, India.
| |
Collapse
|
237
|
Chen G, Chen L, Li X, Mohammadi M. FGF-based drug discovery: advances and challenges. Nat Rev Drug Discov 2025; 24:335-357. [PMID: 39875570 DOI: 10.1038/s41573-024-01125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
The fibroblast growth factor (FGF) family comprises 15 paracrine-acting and 3 endocrine-acting polypeptides, which govern a multitude of processes in human development, metabolism and tissue homeostasis. Therapeutic endocrine FGFs have recently advanced in clinical trials, with FGF19 and FGF21-based therapies on the cusp of approval for the treatment of primary sclerosing cholangitis and metabolic syndrome-associated steatohepatitis, respectively. By contrast, while paracrine FGFs were once thought to be promising drug candidates for wound healing, burns, tissue repair and ischaemic ailments based on their potent mitogenic and angiogenic properties, repeated failures in clinical trials have led to the widespread perception that the development of paracrine FGF-based drugs is not feasible. However, the observation that paracrine FGFs can exert FGF hormone-like metabolic activities has restored interest in these FGFs. The recent structural elucidation of the FGF cell surface signalling machinery and the formulation of a new threshold model for FGF signalling specificity have paved the way for therapeutically harnessing paracrine FGFs for the treatment of a range of metabolic diseases.
Collapse
Affiliation(s)
- Gaozhi Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingfeng Chen
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Moosa Mohammadi
- Institute of Cell Growth Factor, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, Wenzhou, Zhejiang, China.
| |
Collapse
|
238
|
Kavalukas S, Jin A, Babarinde O, Lorkiewicz P, Adhikari J, Xu J, Cai L, DuPre N. Environmental Exposures to Metal Toxins as a Risk Factor for Colorectal Cancer: A Case-Control Study. J Surg Res 2025; 309:269-276. [PMID: 40279913 PMCID: PMC12124944 DOI: 10.1016/j.jss.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the third most common and third most deadly cancer in the United States The role environmental toxins may contribute to CRC incidence is unknown. Cadmium and arsenic are known human carcinogens, although previous data have primarily focused on occupational exposures only. There are no studies on the relationship between environment metal exposures and the incidence of CRC using individual-level measurements from biospecimens. METHODS A pilot case-control study was conducted. Urine and blood specimens were collected. Arsenic and cadmium were measured via inductively coupled plasma mass spectrometry. Logistic regression was used to estimate Odds Ratios of CRC incidence and 95% Confidence Intervals (CI) adjusted for age, gender, family history and smoking or urinary cotinine. Each metal was modeled as a binary variable (high versus low) based on the controls' median value or the limit of detection values. RESULTS Seventy-nine urine and 84 blood specimens were analyzed. Compared to those with low metal levels, the adjusted odds of incident CRC were 1.77 times higher (95% CI: 0.62-5.00), 1.90 times higher (95% CI: 0.68-5.31), and 1.29 times higher (95% CI:0.45-3.72), for those with higher urinary arsenic, urinary cadmium, and blood arsenic, respectively. CONCLUSIONS This is the first study evaluating individual-level measurements of environmental exposures to metal carcinogens and their association with CRC incidence. These pilot results are not statistically significant, although the mildly positive associations may become more profound as recruitment continues. Continued evaluation of environmental toxins and CRC incidence remains warranted.
Collapse
Affiliation(s)
- Sandy Kavalukas
- Division of Colorectal Surgery, Hiram C. Polk Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky; Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville School of Medicine, Louisville, Kentucky.
| | - Allie Jin
- Division of Colorectal Surgery, Hiram C. Polk Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Olufunmilayo Babarinde
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky
| | - Pawel Lorkiewicz
- Bioanalytical Core at the Christina Lee Brown Envirome Institute, University of Louisville, Louisville, Kentucky
| | - Jeevan Adhikari
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky
| | - Jianxiang Xu
- Department of Pediatrics, Pediatric Research Institute, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Lu Cai
- Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville School of Medicine, Louisville, Kentucky; Department of Pediatrics, Pediatric Research Institute, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Natalie DuPre
- Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville School of Medicine, Louisville, Kentucky; Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky
| |
Collapse
|
239
|
Prajapati C, Rai SN, Singh AK, Chopade BA, Singh Y, Singh SK, Haque S, Prieto MA, Ashraf GM. An Update of Fungal Endophyte Diversity and Strategies for Augmenting Therapeutic Potential of their Potent Metabolites: Recent Advancement. Appl Biochem Biotechnol 2025; 197:2799-2866. [PMID: 39907846 PMCID: PMC12065773 DOI: 10.1007/s12010-024-05098-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 02/06/2025]
Abstract
Endophytic fungi represent a significant renewable resource for the discovery of pharmaceutically important compounds, offering substantial potential for new drug development. Their ability to address the growing issue of drug resistance has drawn attention from researchers seeking novel, nature-derived lead molecules that can be produced on a large scale to meet global demand. Recent advancements in genomics, metabolomics, bioinformatics, and improved cultivation techniques have significantly aided the identification and characterization of fungal endophytes and their metabolites. Current estimates suggest there are approximately 1.20 million fungal endophytes globally, yet only around 16% (190,000) have been identified and studied in detail. This underscores the vast untapped potential of fungal endophytes in pharmaceutical research. Research has increasingly focused on the transformation of bioactive compounds by fungal endophytes through chemical and enzymatic processes. A notable example is the anthraquinone derivative 6-O-methylalaternin, whose cytotoxic potential is enhanced by the addition of a hydroxyl group, sharing structural similarities with its parent compound macrosporin. These structure-bioactivity studies open up new avenues for developing safer and more effective therapeutic agents by synthesizing targeted derivatives. Despite the immense promise, challenges remain, particularly in the large-scale cultivation of fungal endophytes and in understanding the complexities of their biosynthetic pathways. Additionally, the genetic manipulation of endophytes for optimized metabolite production is still in its infancy. Future research should aim to overcome these limitations by focusing on more efficient cultivation methods and deeper exploration of fungal endophytes' genetic and metabolic capabilities to fully harness their therapeutic potential.
Collapse
Affiliation(s)
- Chandrabhan Prajapati
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sachchida Nand Rai
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Anurag Kumar Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | | | - Yashveer Singh
- Department of Statistics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Santosh Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
| | - Miguel Angel Prieto
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department. Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004, Ourense, Spain.
| | - Ghulam Md Ashraf
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, 111 Ren'ai road, SIP, Jiangsu Province, Suzhou, 215123, P. R. China.
| |
Collapse
|
240
|
Xiang X, Huang L, Luo W, Qin L, Bian M, Chen W, Han G, Wang N, Mo G, Zhang C, Zhang Y, Yang H, Lu S, Zhang J, Fu T. Neuromuscular electrical stimulation alleviates stroke-related sarcopenia by promoting satellite cells myogenic differentiation via AMPK-ULK1-Autophagy axis. J Orthop Translat 2025; 52:249-264. [PMID: 40342549 PMCID: PMC12059223 DOI: 10.1016/j.jot.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Background Stroke-related sarcopenia can result in muscle mass loss and muscle fibers abnormality, significantly affecting muscle function. The clinical management of stroke-related sarcopenia still requires further research and investigation. This study aims to explore a promising therapy to restore muscle function and promote muscle regeneration in stroke-related sarcopenia, providing a new theory for stroke-related sarcopenia treatment. Methods Stroke-related sarcopenia rat model was established by using permanent middle cerebral artery occlusion (pMCAO) rat and treated with neuromuscular electrical stimulation (NMES). Electrical stimulation (ES) treatment in vitro was mimicked to test the effect of NMES on muscle regeneration in rat skeletal muscle satellite cells (MuSCs). Catwalk, H&E and Masson's trichrome staining, immunofluorescence, transcriptomic analysis, transmission electron microscopy, MuSCs transfection, autophagy flux detection, quantitative real-time PCR analysis, Co-Immunoprecipitation and Western Blot were used to investigate the role of NMES and its mechanism in stroke-related sarcopenia in vivo. Results After NMES treatment, muscle mass and myogenic differentiation were significantly increased in stroke-related sarcopenia rats. The NMES group had more stable gait, neater footprints, higher muscle wet weight, more voluminous morphology and more regenerated muscle fibers. Additionally, ES treatment induced myogenic differentiation in rat MuSCs in vitro. Transcriptomic analysis also showed that "AMPK signaling pathway" was enriched and genes upregulated in ES-treated cells, revealing ES treatment could activate the autophagy in an AMPK-ULK1-dependent mechanism in MuSCs. Besides, it was also founded that infusion of AMPK or ULK1 inhibitor, knockdown of AMPK or ULK1 in MuSCs could block the effect of myotube formation of ES. Conclusion NMES not only restores muscle function but also enhances myogenic activity and muscle regeneration via AMPK-ULK1 autophagy in stroke-related sarcopenia rats. Our study provides a promising strategy for the treatment of stroke-related sarcopenia. The translational potential of this article This study first demonstrates that NMES alleviates stroke-related sarcopenia by promoting MuSCs differentiation through AMPK-ULK1-autophagy axis. The findings reveal a novel therapeutic mechanism, suggesting that NMES can restore muscle function and enhance regeneration in stroke patients. By combining NMES with MuSCs-based therapies, this approach offers a promising strategy for clinical rehabilitation, potentially improving muscle mass and function in stroke survivors. The translational potential lies in its applicability to non-invasive, cost-effective treatments for sarcopenia, enhancing patients' quality of life.
Collapse
Affiliation(s)
- Xingdong Xiang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lei Huang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lieyang Qin
- Department of Cardiovascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guanjie Han
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ning Wang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guokang Mo
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Cheng Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yongxing Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Jian Zhang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Tengfei Fu
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| |
Collapse
|
241
|
Quinlin KA, Beasley JC, Xu X. Metallothionein expression in Eastern mosquitofish (Gambusia holbrooki) exposed to 137Cs and lead: implications for using metallothionein as a metal exposure biomarker. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2025; 44:1322-1333. [PMID: 40037585 DOI: 10.1093/etojnl/vgaf055] [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: 07/25/2024] [Accepted: 02/16/2025] [Indexed: 03/06/2025]
Abstract
Given the range of threats facing aquatic ecosystems, it is critical we have monitoring tools to quickly and efficiently identify stressors within ecosystems. Metallothionein's (MTs) are a group of proteins widely used as biomarkers of metal exposure; however, their suitability has been questioned due to their influenceable nature and ability to be induced by various stressors. Through our study, we sought to (1) evaluate induction of MT in eastern mosquitofish (Gambusia holbrooki) exposed to a radionuclide (137Cs) and metal (Pb), (2) assess the influence of in situ 137Cs pre-exposure on Pb-induced MT expression, and (3) quantify changes in MT concentrations during Pb depuration. We conducted our experiment in three phases using mosquitofish as a model species: Phase I-pre-exposure to 137Cs in the field, Phase II-exposure to Pb in the lab immediately following 137Cs exposure, and Phase III-depuration in the laboratory. We quantified MT concentrations over time and assessed differences among treatments. We did not find in situ exposure of mosquitofish to 137Cs to induce MT, nor did pre-exposure to 137Cs appear to influence induction of MT following Pb exposure. Metallothionein was rapidly induced in mosquitofish following Pb exposure; however, MT induction was also observed in control fish. Additionally, there was no apparent relationship between MT and Pb body burdens. During depuration, we found MT concentrations to be highly variable among fish demonstrating no clear response. Our study demonstrates the high variability exhibited by MT, the need for further validation of MT as a metal exposure biomarker, and the value in additional research on these proteins under varying exposure scenarios, particularly environmentally realistic concentrations.
Collapse
Affiliation(s)
- Kathryn A Quinlin
- Warnell School of Forestry and Natural Resources, University of Georgia, Athens, GA, United States
- Savannah River Ecology Lab, University of Georgia, Aiken, SC, United States
| | - James C Beasley
- Warnell School of Forestry and Natural Resources, University of Georgia, Athens, GA, United States
- Savannah River Ecology Lab, University of Georgia, Aiken, SC, United States
| | - Xiaoyu Xu
- Warnell School of Forestry and Natural Resources, University of Georgia, Athens, GA, United States
- Savannah River Ecology Lab, University of Georgia, Aiken, SC, United States
| |
Collapse
|
242
|
Shen Q, Yang Z, Hu C, Liu Y, Zhao L, Li C, Ma Y, Bian H. Non-starch polysaccharides and health: gut-target organ axis influencing obesity. Food Sci Biotechnol 2025; 34:1771-1788. [PMID: 40196321 PMCID: PMC11972281 DOI: 10.1007/s10068-024-01745-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 04/09/2025] Open
Abstract
Obesity is recognized as a global epidemic that can result in changes in the human body and metabolism. Accumulating evidence indicates that gut microbiota (GM) can affect the development of obesity. The GM not only plays a crucial role in digesting and absorbing nutrients, but also in maintaining the overall health of the host. Dietary supplements such as non-starch polysaccharides are mainly fermented by the GM in the colon. Recent findings suggest that shaping the GM through the prebiotic function of non-starch polysaccharides may be a viable strategy against obesity. In this paper, the effects of non-starch polysaccharides on host health, together with their prebiotic function influencing the GM to control obesity via the gut-target organ axis, are reviewed. Potential perspectives of non-starch polysaccharides exhibiting anti-obesity effects via the gut-target organ axis are proposed for future research. Graphical abstract
Collapse
Affiliation(s)
- Qingshan Shen
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Zhuan Yang
- School of Life Science and Agricultural Engineering, Nanyang Normal University, Wolong Road 1638, Nanyang, 473061 China
| | - Chengzhi Hu
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071000 China
| | - Yilin Liu
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Lei Zhao
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Cuicui Li
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Yanli Ma
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Hua Bian
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| |
Collapse
|
243
|
Mansouri F, Seyed Mohammadzad M. Bioinformatics analyses of potential microRNAs and their target genes in myocardial infarction patients with diabetes. Diab Vasc Dis Res 2025; 22:14791641251335925. [PMID: 40326247 PMCID: PMC12059454 DOI: 10.1177/14791641251335925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 01/24/2025] [Accepted: 04/03/2025] [Indexed: 05/07/2025] Open
Abstract
ObjectivePatients with diabetes are 3-5 times higher at risk for cardiovascular diseases and myocardial infarction (MI). There is a need to find miRNAs and other target genes to reduce mortality rates. The current study aims to find potential miRNAs and target genes among MI patients, MI patients with pre-diabetes (metformin non-users), and MI patients with diabetes (metformin users).MethodThe candidate miRNAs were identified by microarray profiling, and their differential expression was evaluated through real-time polymerase chain reaction (RT-PCR) in control and patient groups. The potential targets for miR-1 and miR-133a were retrieved from the TargetScan, miRWalk, and miRDB databases. The sensitivity and specificity of miRNAs were assessed using receiver operating characteristic (ROC) curve analyses.ResultsMicroarray profiling identified 16 miRNAs with significantly altered expression in all MI patient groups compared with healthy controls. According to this data, two miR-1 and miR-133a (with a high ratio) were selected for further verification. All patient groups exhibited a significant increase in the expression levels of miR-1 and miR-133a. Also, miR-1 and miR-133a levels were lower in metformin-user patients than in non-user patients (p < 0.05). Moreover, interleukins, growth factors, and other related genes were identified as potential targets for miR-1 and miR-133a. The ROC area under the curve (AUC) was 0.973 (95% CI: 0.718-0.884) for circulating miR-1, and 0.969 (95% CI: 0.723-0.876) for miR-133a in patients with diabetes (p < 0.001).ConclusionPrediction of miRNA profiles and network of target genes are valuable in the early diagnosis of MI in individuals without and with diabetes. Metformin treatment is associated with lower expression of MI-related miRNAs, suggesting a potential mechanism for cardiac protection by this agent.
Collapse
Affiliation(s)
- Fatemeh Mansouri
- Department of Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Iran
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Iran
| | | |
Collapse
|
244
|
Ren X, Yang P, Sun L, Jia Y, Zhong C, Zhang J, Zhang Q, Xu T, Zheng X, Zhang Y. High plasma fibroblast growth factor 19 is associated with improved prognosis in patients with acute ischemic stroke. Clin Nutr 2025; 48:16-24. [PMID: 40117962 DOI: 10.1016/j.clnu.2025.03.013] [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: 12/31/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE To prospectively investigate the relationships between plasma fibroblast growth factor 19 (FGF-19) and clinical outcomes in patients with acute ischemic stroke. METHODS Plasma FGF-19 levels at baseline were measured for 3048 patients with ischemic stroke, and all patients were followed up at one year after stroke onset. The primary outcome was a combination of death and major disability (modified Rankin Scale score of ≥3) at one year after stroke onset, and secondary outcomes included major disability, death, recurrent stroke, vascular events and the combination of death and vascular events. RESULTS During the 1-year of follow-up, 682 (22.38 %) patients experienced the primary outcome; 503 had a major disability and 179 died. After multivariate adjustment, higher plasma FGF-19 was significantly associated with decreased risk of the primary outcome (odds ratio = 0.48, 95 % confidence interval = 0.36-0.66). Each 1-SD increase of log-transformed FGF-19 (0.93 pg/mL) was associated with 20 % decreased risk of the primary outcome. The addition of FGF-19 to the conventional risk factors significantly improved prediction of the primary outcome in ischemic stroke patients (net reclassification index = 25.10 %, p < 0.001; integrated discrimination improvement = 1.25 %, p < 0.001). Furthermore, patients with both high FGF-19 (≥573.7 pg/mL) and low FGF-21 (<740.1 pg/mL) levels had the lowest incidences of all study outcomes. CONCLUSIONS High plasma FGF-19 levels were associated with improved prognosis in patients with acute ischemic stroke, suggesting that FGF-19 may be a potential biomarker of good prognosis for ischemic stroke.
Collapse
Affiliation(s)
- Xiao Ren
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yiming Jia
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-Communicable Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jing Zhang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qilu Zhang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-Communicable Diseases, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xiaowei Zheng
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-Communicable Diseases, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
245
|
Bellver‐Sanchis A, Ribalta‐Vilella M, Irisarri A, Gehlot P, Choudhary BS, Jana A, Vyas VK, Banerjee DR, Pallàs M, Guerrero A, Griñán‐Ferré C. G9a an Epigenetic Therapeutic Strategy for Neurodegenerative Conditions: From Target Discovery to Clinical Trials. Med Res Rev 2025; 45:985-1015. [PMID: 39763018 PMCID: PMC11976383 DOI: 10.1002/med.22096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 04/09/2025]
Abstract
This review provides a comprehensive overview of the role of G9a/EHMT2, focusing on its structure and exploring the impact of its pharmacological and/or gene inhibition in various neurological diseases. In addition, we delve into the advancements in the design and synthesis of G9a/EHMT2 inhibitors, which hold promise not only as a treatment for neurodegeneration diseases but also for other conditions, such as cancer and malaria. Besides, we presented the discovery of dual therapeutic approaches based on G9a inhibition and different epigenetic enzymes like histone deacetylases, DNA methyltransferases, and other lysine methyltransferases. Hence, findings offer valuable insights into developing novel and promising therapeutic strategies targeting G9a/EHMT2 for managing these neurological conditions.
Collapse
Affiliation(s)
- Aina Bellver‐Sanchis
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Marta Ribalta‐Vilella
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Alba Irisarri
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Pinky Gehlot
- Department of Pharmaceutical ChemistryInstitute of PharmacyNirma UniversityAhmedabadIndia
| | - Bhanwar Singh Choudhary
- Department of PharmacyCentral University of RajasthanAjmerIndia
- Drug Discovery and Development Centre (H3D)University of Cape TownRondeboschSouth Africa
| | - Abhisek Jana
- Department of ChemistryNational Institute of Technology DurgapurDurgapurIndia
| | - Vivek Kumar Vyas
- Department of Pharmaceutical ChemistryInstitute of PharmacyNirma UniversityAhmedabadIndia
| | - Deb Ranjan Banerjee
- Department of ChemistryNational Institute of Technology DurgapurDurgapurIndia
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
- Instituto de Salud Carlos III, Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Ana Guerrero
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Christian Griñán‐Ferré
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
- Instituto de Salud Carlos III, Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| |
Collapse
|
246
|
Huang M, Chang J, Liu Y, Yin J, Zeng X. Apelin/APJ alleviates diabetic nephropathy by improving glomerular endothelial cells dysfunction via SIRT3‑KLF15. Mol Med Rep 2025; 31:122. [PMID: 40052569 PMCID: PMC11920778 DOI: 10.3892/mmr.2025.13487] [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: 10/17/2024] [Accepted: 01/07/2024] [Indexed: 03/21/2025] Open
Abstract
Glomerular basement membrane (GBM) thickening, the earliest morphological change of diabetic nephropathy (DN), is related to glomerular endothelial cells (GECs) dysfunction which increase extracellular matrix (ECM) synthesizing. Apelin, the endogenous ligand for apelin/apelin receptor (APJ), is reported to alleviate endothelial cell dysfunction in DN. Therefore, it was hypothesized that apelin/APJ reduced GBM thickening by decreasing the synthesis of ECM in GECs. The results showed that apelin reduced glomerular fibrosis and GBM thickening by decreasing the expression of laminin and collagen IV in diabetic mice, which were cancelled following APJ knockout in GECs. Furthermore, apelin/APJ inhibited the synthesis of laminin and collagen IV in GECs by increasing the expression and activity of SIRT3, which promoted KLF15 deacetylation and translocation into nucleus. In conclusion, apelin/APJ reduced GBM thickening in diabetes mellitus by preventing laminin and collagen IV synthesizing via SIRT3‑KLF15 pathway in GECs.
Collapse
Affiliation(s)
- Mingcong Huang
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, P.R. China
| | - Jing Chang
- Department of Physiology, Beijing You An Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Yu Liu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, P.R. China
| | - Jiming Yin
- Beijing Institute of Hepatology, Beijing You An Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
247
|
Zhao Y, Qin G, Fan W, Zhang Y, Peng H. TF and TFRC regulate ferroptosis in swine testicular cells through the JNK signaling pathway. Int J Biol Macromol 2025; 307:142369. [PMID: 40120870 DOI: 10.1016/j.ijbiomac.2025.142369] [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: 01/08/2025] [Revised: 02/23/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Transferrin (TF) is a serum glycoprotein that plays a critical role in iron metabolism and typically functions through binding to its transferrin receptor (TFRC). TF is also considered a key indicator of sperm quality and, together with TFRC, plays a critical role in regulating spermatogenesis. This study aimed to explore the effects of increased TF and TFRC expression on ferroptosis in swine testicular cells (ST cells). Our findings revealed that the overexpression of either TF or TFRC diminishes ST cell viability, increases cytotoxicity, intensifies oxidative stress damage, decreases mitochondrial activity, and promotes ferroptosis. Transcriptomic analysis suggested that TF and TFRC may influence ST cells through the MAPK signaling pathway. Subsequent experiments revealed that inhibiting the JNK signaling pathway within the MAPK pathway improved mitochondrial activity, reduced oxidative stress damage, and mitigated ferroptosis progression. Moreover, we discovered that TF and TFRC might regulate cellular oxidative phosphorylation via the JNK signaling pathway. In conclusion, increased expression of TF or TFRC increases the sensitivity of ST cells to ferroptosis and modulates mitochondrial DNA transcription and energy metabolism through the JNK signaling pathway. These findings could offer potential therapeutic targets for addressing reproductive toxicity associated with ferroptosis.
Collapse
Affiliation(s)
- Yuanjie Zhao
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; College of Life and Health, Hainan University, Haikou 570228, China
| | - Ge Qin
- College of Animal Science and Technology, Southwest University, Chongqing 404100, China
| | - Weiqin Fan
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Yanyan Zhang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Hui Peng
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China.
| |
Collapse
|
248
|
Hou HT, Chen HX, Wang ZQ, Xi L, Wang J, Yang Q, He GW. Arterial and venous grafting biomaterials in coronary Surgery: Integrative multi-omics approach reveals ECM-PI3K-Akt pathway as Key Regulator of different patency. CHEMICAL ENGINEERING JOURNAL 2025; 511:161829. [DOI: 10.1016/j.cej.2025.161829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
|
249
|
Su J, Zhang Y, Wang X, Hu X, Zhou K, Zhu H, Liu E, Liu S. Huangkui capsules regulate tryptophan metabolism to improve diabetic nephropathy through the Keap1/Nrf2/HO-1 pathway. Front Pharmacol 2025; 16:1535352. [PMID: 40371325 PMCID: PMC12075421 DOI: 10.3389/fphar.2025.1535352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
Background Diabetic nephropathy (DN) is a serious complication of diabetes and one of the leading causes of end-stage renal disease. Huangkui capsule (HKC), a traditional Chinese patent medicine, is widely used in clinical practice for the treatment of chronic glomerulonephritis. However, the therapeutic effects and underlying mechanisms of HKC in DN remain poorly understood. Methods DN was induced in db/db mice, which were randomly divided into the DN, HKC-L, HKC-H and IRB groups, and db/m mice served as the Control group. Biochemical indices of blood and urine samples from the mice were measured, and HE staining, Masson staining and PAS staining were used to verify the anti-DN effect of HKC. The levels of ROS and the expression of Nrf2 pathway-related proteins and mRNAs were detected. Metabonomic analysis was used to investigate the role of tryptophan metabolism in the regulation of DN by HKC. HK-2 cells were used to establish a model of high-glucose (HG) injury in vitro, and HKC treatment was given for supplementary verification. Sarpogrelate hydrochloride (SH) combined with HKC, a 5-HT2AR inhibitor, was used to verify the effect of the 5-HT pathway in an in vitro model. Results Treatment with HKC significantly inhibited the increase in blood glucose and Urinary albumin/creatinine ratio (UACR), improved kidney injury signs in mice, reduced the level of ROS and improved oxidative stress injury through the Keap1/ Nrf2/HO-1 pathway. Metabonomic analysis revealed that tryptophan metabolism is involved in the process by which HKC improves DN, and HKC can regulate the 5-HT pathway to improve the renal injury by oxidative stress regulation. HKC treatment also significantly improved the renal and oxidative stress injuries in HG HK-2 cell model through the Nrf2 pathway in vitro. SH administration revealed that inhibiting 5-HT2AR could significantly inhibit the synthesis of 5-HT and improve the renal injury induced by HG. Conclusion Our study demonstrate that HKC can inhibit kidney injury and oxidative stress injury in db/db mice and HK-2 cells by regulating tryptophan metabolism and the Keap1/Nrf2/HO-1 pathway, which provides new insight for the clinical use of HKC for treatment of DN.
Collapse
Affiliation(s)
- Jiayu Su
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ying Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuan Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaochao Hu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ke Zhou
- College of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Huimin Zhu
- College of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ehu Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shijia Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
250
|
Muttiah B, Hanafiah A. Gut Microbiota and Cardiovascular Diseases: Unraveling the Role of Dysbiosis and Microbial Metabolites. Int J Mol Sci 2025; 26:4264. [PMID: 40362500 PMCID: PMC12072866 DOI: 10.3390/ijms26094264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure (HF), hypertension, myocardial infarction (MI), and atherosclerosis, are increasingly linked to gut microbiota dysbiosis and its metabolic byproducts. HF, affecting over 64 million individuals globally, is associated with systemic inflammation and gut barrier dysfunction, exacerbating disease progression. Similarly, hypertension and MI correlate with reduced microbial diversity and an abundance of pro-inflammatory bacteria, contributing to vascular inflammation and increased cardiovascular risk. Atherosclerosis is also influenced by gut dysbiosis, with key microbial metabolites such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs) playing crucial roles in disease pathogenesis. Emerging evidence highlights the therapeutic potential of natural compounds, including flavonoids, omega-3 fatty acids, resveratrol, curcumin, and marine-derived bioactives, which modulate the gut microbiota and confer cardioprotective effects. These insights underscore the gut microbiota as a critical regulator of cardiovascular health, suggesting that targeting dysbiosis may offer novel preventive and therapeutic strategies. Further research is needed to elucidate underlying mechanisms and optimize microbiome-based interventions for improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- GUT Research Group, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|