1
|
Ginet V, Depierre P, Puyal J. Neuronal autosis: the self-destructive side of autophagy involved in hypoxic-ischemic neuronal death. Neural Regen Res 2025; 20:3517-3518. [PMID: 39589723 PMCID: PMC11974643 DOI: 10.4103/nrr.nrr-d-24-00831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/13/2024] [Accepted: 10/11/2024] [Indexed: 11/27/2024] Open
Affiliation(s)
- Vanessa Ginet
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland
| | - Pauline Depierre
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
- CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
2
|
Li C, Ji H, Zhuang S, Xie X, Cui D, Zhang C. Update on the correlation between mitochondrial function and osteonecrosis of the femoral head osteocytes. Redox Rep 2025; 30:2491846. [PMID: 40249372 PMCID: PMC12010656 DOI: 10.1080/13510002.2025.2491846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025] Open
Abstract
Mitochondrial health is maintained in a steady state through mitochondrial dynamics and autophagy processes. Recent studies have identified healthy mitochondria as crucial regulators of cellular function and survival. This process involves adenosine triphosphate (ATP) synthesis by mitochondrial oxidative phosphorylation (OXPHOS), regulation of calcium metabolism and inflammatory responses, and intracellular oxidative stress management. In the skeletal system, they participate in the regulation of cellular behaviors and the responses of osteoblasts, osteoclasts, chondrocytes, and osteocytes to external stimuli. Indeed, mitochondrial damage or dysfunction occurs in the development of a few bone diseases. For example, mitochondrial damage may lead to an imbalance in osteoblasts and osteoclasts, resulting in osteoporosis, osteomalacia, or poor bone production, and chondrocyte death and inflammatory infiltration in osteoarthritis are the main causes of cartilage degeneration due to mitochondrial damage. However, the opposite exists for osteosarcoma, where overactive mitochondrial metabolism is able to accelerate the proliferation and migration of osteosarcoma cells, which is a major disease feature. Bone is a dynamic organ and osteocytes play a fundamental role in all regions of bone tissue and are involved in regulating bone integrity. This review examines the impact of mitochondrial physiological function on osteocyte health and summarizes the microscopic molecular mechanisms underlying its effects. It highlights that targeted therapies focusing on osteocyte mitochondria may be beneficial for osteocyte survival, providing a new insight for the diagnosis, prevention, and treatment of diseases associated with osteocyte death.
Collapse
Affiliation(s)
- Chengming Li
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Hangyu Ji
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Suyang Zhuang
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Xinhui Xie
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Daping Cui
- Department of Orthopedics, Shenzhen Bao’an District Central Hospital, Shenzhen, People’s Republic of China
| | - Cong Zhang
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| |
Collapse
|
3
|
Sun S, Arif Aslam M, Ma EB, Lee G, Muhammad Ahmad Javaid H, Yoon S, Huh JY. Activation of CXCR7 exerts an inhibitory effect on adipogenesis through regulation of β-arrestin2/Wnt and AKT signalling. Adipocyte 2025; 14:2490258. [PMID: 40302245 PMCID: PMC12045560 DOI: 10.1080/21623945.2025.2490258] [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: 08/27/2024] [Revised: 01/22/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025] Open
Abstract
CXCR7, an alternative receptor for the inflammatory chemokine SDF-1, is involved in cell proliferation and migration. Recent studies have reported that CXCR7 also plays a role in adipose tissue. However, evidence regarding the role of CXCR7 and its ligands in adipocyte differentiation is limited. In this study, we aimed to elucidate changes in CXCR7 expression during adipocyte differentiation and the role of the SDF-1/CXCR7/CXCR4 axis in adipogenesis using recombinant SDF-1, the CXCR7 ligand CCX771, and small interfering RNAs. The results indicated that the levels of SDF-1 and its receptors, CXCR7 and CXCR4, decreased during the early stages of adipogenesis. Treatment with recombinant SDF-1 and CCX771 inhibited adipogenesis and lipid accumulation by inducing β-arrestin2, Wnt expression, and AKT phosphorylation and downregulating C/EBPα, PPARγ, and FABP4 expression. In contrast, knockdown of SDF-1 and CXCR7 in preadipocytes downregulated the β-arrestin2/Wnt and AKT pathway, leading to the induction of adipogenesis. Meanwhile, knockdown of CXCR4 had no significant effect. In mice, basal gene expression levels of SDF-1 and CXCR7 were higher in the stromal vascular fraction compared to mature adipocytes and were significantly upregulated by a high-fat diet. Our results provide new insights into the local role of the SDF-1-CXCR7 axis in adipocytes and offer additional benefits for the prevention of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Shiyue Sun
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Gahui Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hafiz Muhammad Ahmad Javaid
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Somy Yoon
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Zhang Y, Wei Y, Han X, Shi L, Yu H, Ji X, Gao Y, Gao Q, Zhang L, Duan Y, Li W, Yuan Y, Shi J, Cheng L, Li Y. Faecalibacterium prausnitzii prevents age-related heart failure by suppressing ferroptosis in cardiomyocytes through butyrate-mediated LCN2 regulation. Gut Microbes 2025; 17:2505119. [PMID: 40364435 PMCID: PMC12080280 DOI: 10.1080/19490976.2025.2505119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/20/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025] Open
Abstract
Aging is a primary driver of the escalating prevalence of heart failure (HF). Age-associated gut microbiota dysbiosis has been implicated in various age-related diseases, yet its role in age-related HF remains largely unexplored. In this study, we sought to explore the potential link between age-related gut microbiota alterations and HF in the elderly. We analyzed a publicly available single-cell sequencing dataset, which revealed markedly increased ferroptosis activity in cardiac myocytes of elderly individuals compared to their younger counterparts. Notably, treatment with the ferroptosis inhibitor, ferrostatin-1, mitigated cardiac ferroptosis and prevented cardiac dysfunction in aging rats. Furthermore, fecal microbiota transplantation from elderly HF patients significantly increased cardiac ferroptosis activity and induced cardiac dysfunction in healthy recipient rats. Integrated 16S rRNA sequencing and PCR quantification revealed a marked depletion of Faecalibacterium prausnitzii (F. prausnitzii) in elderly individuals, with a more pronounced decline in elderly patients with HF. Oral administration of F. prausnitzii or its metabolite butyrate effectively attenuated age-related HF through inhibiting ferroptosis. Additionally, gene-editing techniques were employed to generate F. prausnitzii BCoAT mutant deficient in butyrate production. Intriguingly, the protective effect was lost in the butyrate-deficient F. prausnitzii strain. Mechanistically, butyrate reduced intracellular iron accumulation and suppressed ferroptosis by downregulating LCN2 expression in senescent cardiomyocytes. Our findings highlight the critical role of aged microbiota-induced ferroptosis in HF and propose F. prausnitzii or butyrate may serve as potential targets for the prevention and treatment of age-related HF.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Wei
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xuejie Han
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ling Shi
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hui Yu
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuanrui Ji
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yunlong Gao
- Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin, China
| | - Qianhui Gao
- Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin, China
| | - Linwei Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yu Duan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenpeng Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yue Yuan
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jing Shi
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
Ren X, Zhao L, Hao Y, Huang X, Lv G, Zhou X. Copper-instigated modulatory cell mortality mechanisms and progress in kidney diseases. Ren Fail 2025; 47:2431142. [PMID: 39805816 PMCID: PMC11734396 DOI: 10.1080/0886022x.2024.2431142] [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: 02/28/2024] [Revised: 06/23/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
Copper is a vital cofactor in various enzymes, plays a pivotal role in maintaining cell homeostasis. When copper metabolism is disordered and mitochondrial dysfunction is impaired, programmed cell death such as apoptosis, paraptosis, pyroptosis, ferroptosis, cuproptosis, autophagy and necroptosis can be induced. In this review, we focus on the metabolic mechanisms of copper. In addition, we discuss the mechanism by which copper induces various programmed cell deaths. Finally, this review examines copper's involvement in prevalent kidney diseases such as acute kidney injury and chronic kidney disease. The findings indicate that the use of copper chelators or plant extracts can mitigate kidney damage by reducing copper accumulation, offering novel insights into the pathogenesis and treatment strategies for kidney diseases.
Collapse
Affiliation(s)
- Xiya Ren
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Limei Zhao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yajie Hao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiu Huang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guangna Lv
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
García-Juan M, Villa M, Benito-Cuesta I, Ordóñez-Gutiérrez L, Wandosell F. Reassessing the AMPK-MTORC1 balance in autophagy in the central nervous system. Neural Regen Res 2025; 20:3209-3210. [PMID: 39715086 PMCID: PMC11881726 DOI: 10.4103/nrr.nrr-d-24-00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Marta García-Juan
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mario Villa
- Fisiología Animal, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Irene Benito-Cuesta
- Department of Clinical Neuroscience, CMM Karolinska Universitetssjukhuset Solna, Stockholm, Sweden
| | - Lara Ordóñez-Gutiérrez
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
7
|
Nakhaei A, Marzoughi S, Ghoflchi S, Hosseini H, Afshari AR, Jalili-Nik M, Kesharwani P, Sahebkar A. An exploration of molecular signaling in drug reprocessing for Oral Squamous Cell Carcinoma. Eur J Med Chem 2025; 295:117816. [PMID: 40466285 DOI: 10.1016/j.ejmech.2025.117816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2025] [Revised: 05/14/2025] [Accepted: 05/25/2025] [Indexed: 06/11/2025]
Abstract
The unique characteristics of cancer are crucial for comprehending the processes underlying cancer initiation, development, and maintenance. These hallmarks guide the development of novel therapeutic strategies aimed at fundamental traits of cancer, resulting in more targeted therapies with the possibility for sustained effectiveness and minimized adverse effects. Drug repurposing, a novel approach that leverages the known safety and pharmacological properties of existing drugs, has surfaced as a viable alternative to traditional drug development. This method expedites the timescale for introducing novel medicines into clinical practice, often demonstrating reduced failure rates in clinical trials. Recent data substantiates the therapeutic efficacy of many repurposed medications in the management of oral squamous cell carcinomas (OSCC), a highly aggressive and treatment-resistant malignancy. Prominent instances include metformin, phenformin, propranolol, acetylsalicylic acid, celecoxib, itraconazole, statins, dihydroartemisinin, and methotrexate. These pharmaceuticals demonstrated diverse anticancer actions, rendering them valuable tools in the therapy of OSCC. This review provides a comprehensive overview of molecular signaling in the reprocessing of drugs for OSCC.
Collapse
Affiliation(s)
- Ali Nakhaei
- Student Research Committee, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Sarah Marzoughi
- Student Research Committee, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Sahar Ghoflchi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Basic Sciences, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; Applied Biomedical Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Sharma C, Kim S, Eo H, Kim SR. Recovery of the injured neural system through gene delivery to surviving neurons in Parkinson's disease. Neural Regen Res 2025; 20:2855-2861. [PMID: 39610091 PMCID: PMC11826474 DOI: 10.4103/nrr.nrr-d-24-00724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/22/2024] [Accepted: 09/11/2024] [Indexed: 11/30/2024] Open
Abstract
A critical unaddressed problem in Parkinson's disease is the lack of therapy that slows or hampers neurodegeneration. While medications effectively manage symptoms, they offer no long-term benefit because they fail to address the underlying neuronal loss. This highlights that the elusive goals of halting progression and restoring damaged neurons limit the long-term impact of current approaches. Recent clinical trials using gene therapy have demonstrated the safety of various vector delivery systems, dosages, and transgenes expressed in the central nervous system, signifying tangible and substantial progress in applying gene therapy as a promising Parkinson's disease treatment. Intriguingly, at diagnosis, many dopamine neurons remain in the substantia nigra, offering a potential window for recovery and survival. We propose that modulating these surviving dopamine neurons and axons in the substantia nigra and striatum using gene therapy offers a potentially more impactful therapeutic approach for future research. Moreover, innovative gene therapies that focus on preserving the remaining elements may have significant potential for enhancing long-term outcomes and the quality of life for patients with Parkinson's disease. In this review, we provide a perspective on how gene therapy can protect vulnerable elements in the substantia nigra and striatum, offering a novel approach to addressing Parkinson's disease at its core.
Collapse
Affiliation(s)
- Chanchal Sharma
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
| | - Sehwan Kim
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
| | - Hyemi Eo
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
| | - Sang Ryong Kim
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| |
Collapse
|
9
|
Tang Y, Wu F, Xing W, Zhao C, Pan Y, Han J, Wu G, Huang Y. Morphology dependence of zeolitic imidazolate framework-67 nanoreactor for carbon dioxide reduction and hexavalent chromium immobilisation. J Colloid Interface Sci 2025; 693:137649. [PMID: 40262211 DOI: 10.1016/j.jcis.2025.137649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
Energy conversion and pollutant remediation are essential means of environmental protection. Zeolitic imidazolate framework-67 (ZIF-67) is an ideal catalyst for these processes. In this study, four morphologies of ZIF-67 were synthesised for the photocatalytic conversion of carbon dioxide (CO2) and adsorption of hexavalent chromium (Cr(VI)). Under light irradiation, rhombic ZIF-67 (ZIF-R) converted carbon dioxide to carbon monoxide (CO) and methane (CH4), with production rates of 1182.62 and 27.21 μmol g-1 h-1, respectively. Furthermore, comprehensive experiments were conducted to examine the effects of reaction duration, pH, starting concentration, coexisting ions and temperature, along with corresponding kinetic and thermodynamic analyses. Results demonstrated that ZIF-R exhibited outstanding Cr(VI) adsorption capacity (46.87 mg g-1). After four cycles of adsorption and photocatalytic experiments, ZIF-R maintained excellent reusability. Thus, ZIF-R is an efficient photocatalyst and stable adsorbent, offering a promising solution for mitigating greenhouse gas emissions and removing environmental pollutants.
Collapse
Affiliation(s)
- Yonggong Tang
- College of Ecology and Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Fan Wu
- College of Ecology and Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Weinan Xing
- College of Ecology and Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; National Positioning Observation Station of Hung-tse Lake Wetland Ecosystem in Jiangsu Province, Hongze 223100, China
| | - Chunyu Zhao
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, China
| | - Yuwei Pan
- College of Ecology and Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China
| | - Jiangang Han
- College of Ecology and Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; National Positioning Observation Station of Hung-tse Lake Wetland Ecosystem in Jiangsu Province, Hongze 223100, China
| | - Guangyu Wu
- College of Ecology and Environment, Co-Innovation Center for the Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China; National Positioning Observation Station of Hung-tse Lake Wetland Ecosystem in Jiangsu Province, Hongze 223100, China.
| | - Yudong Huang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
10
|
Mayyas F, Omeish A. Comparison of the cardioprotective effects of liraglutide, dapagliflozin and their combination in a rat model of diabetes induced by streptozotocin. Life Sci 2025; 375:123721. [PMID: 40389022 DOI: 10.1016/j.lfs.2025.123721] [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: 02/23/2025] [Revised: 05/02/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP-1) receptor agonists positively affect diabetic cardiac complications. AIM To evaluate and compare the impact of liraglutide, dapagliflozin, and their combination on cardiac biomarkers of inflammation, oxidative stress, and fibrosis in a rat model of streptozotocin (STZ)-induced diabetes. METHODS Adult male Wistar rats were assigned into five groups (15-17 rats/group): control rats, diabetic rats (DM, single 50 mg/kg STZ intraperitoneally (IP)), diabetic rats treated with dapagliflozin (Dapa, 1 mg/kg by oral gavage), diabetic rats treated with liraglutide (Lira), 0.4 mg/kg subcutaneously (SC), and diabetic rats treated with both medications (Dapa+Lira) for 8 weeks. Cardiac biomarkers of inflammation, oxidative stress, and fibrosis were evaluated. RESULTS Dapagliflozin and/or liraglutide treatment lowered glucose levels, mostly in the combination group. Diabetes increased heart/body weight ratio, which was normalized by all treatments. DM increased cardiac inflammatory and oxidant markers, including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), endothelin-1 (ET-1), myeloperoxidase (1), plasminogen activator inhibitor-2 (PAI-2), total nitrite, and thiobarbituric acid reactive substances (TBARS). Dapagliflozin normalized inflammatory markers levels, but combination with Lira added no benefit, except for PAI-2. Dapagliflozin normalized total nitrite and TBARS levels. Combining treatments further decreased nitrite and TBARS levels and normalized cardiac SOD activity. Both dapagliflozin and the combination normalized cardiac fibrosis and platelet-derived growth factor-BB (PDGF-BB) levels. CONCLUSION Dapagliflozin reduced cardiac fibrosis, and attenuated oxidative stress, and inflammation more effectively than liraglutide. Combining treatments improved oxidative status. Our findings support using dapagliflozin to prevent cardiovascular diseases more than liraglutide.
Collapse
Affiliation(s)
- Fadia Mayyas
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| | - Anood Omeish
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| |
Collapse
|
11
|
Bhardwaj P, Maruthi M. Hyperglycaemia-induced fibrotic and inflammatory gene expression alterations in lung epithelial cells: Implications for pulmonary fibrosis development. Gene 2025; 959:149520. [PMID: 40254080 DOI: 10.1016/j.gene.2025.149520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/22/2025]
Abstract
Hyperglycaemia has a significant long-term impact on multiple organ systems, including renal, cardiovascular, central nervous, hepatic and ocular systems, leading to the gradual loss of their functional abilities. Numerous studies have elucidated the pathophysiology, etiology, and consequences of hyperglycaemia on these organs. The pulmonary system is also considered as a target of hyperglycaemia, several factors cause lung injury which leads to the development of pulmonary fibrosis, a chronic fibrotic disease with usual interstitial pneumonia patterns. Nevertheless, the effects of hyperglycaemia on the development of pulmonary fibrosis remain poorly understood. We intend to understand the cellular and morphological changes, and the progression of fibrosis in lung epithelial cells subjected to hyperglycaemia. Our experimental data indicate that hyperglycaemia induces fibrotic and inflammatory alterations in cultured lung epithelial cells. These alterations are facilitated by the upregulation of genes related to fibrosis and inflammation, promoting cell proliferation and migration. Further research is required to comprehensively elucidate the impact of hyperglycaemia during lung injury progression of fibrosis, these findings may reveal novel mechanisms that may help in the assessment and treatment of lung ailments in people with hyperglycaemia.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Biochemistry, School of Interdisciplinary and Applied Sciences, Central University of Haryana
| | - Mulaka Maruthi
- Department of Biochemistry, School of Interdisciplinary and Applied Sciences, Central University of Haryana.
| |
Collapse
|
12
|
Silva-Palacios A, Zúñiga-Muñoz AM, Soria-Castro E, Álvarez-León E, Nieto M, Navarrete-Anastasio G, Carbó R, García-Niño WR, López-Cervantes SP, Salas-Venegas V, Flores-Torres RP, Luna-López A, Zazueta C, Königsberg M. Cardioprotective effect of senotherapy in chronically obese middle-aged female rats may be mediated by a MERCSs/Nrf2 interaction. J Nutr Biochem 2025; 142:109923. [PMID: 40250489 DOI: 10.1016/j.jnutbio.2025.109923] [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: 12/18/2024] [Revised: 03/16/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Hypercaloric intake promotes the development of obesity, a risk factor for cardiovascular disease (CVD). In recent years, it has been suggested that senescent cells have negative implications for the outcome of these chronic pathologies, and senotherapy has emerged as a novel intervention to reduce damage to the organism. However, it is unclear whether the accumulation of senescent cells induces alterations at the cardiac level in rats fed a hypercaloric diet (HD) and if the use of senotherapeutics can reverse it. To address this question, we used middle-aged female rats fed HD from 21 days to 15 months of age. Under our experimental conditions, rats exhibited cardiac hypertrophy and fibrosis, accumulation of senescent cells, changes in mitochondrial morphology, and oxidative stress. Rats were treated for 2 months with senolytic (dasatinib + quercetin, DQ) or senomorphic (sulforaphane, SFN) agents. Interestingly, the HD rats showed cardiac improvement after the treatment. Our data suggest a possible link mechanism between Nrf2 activation and mitochondria-endoplasmic reticulum contact sites (MERCSs) preservation, activated by SFN rather than by the DQ combination, which allowed cardiac structure maintenance in HD rats decreasing the harmful effects of senescent cells.
Collapse
Affiliation(s)
- Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| | - Alejandra María Zúñiga-Muñoz
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Elizabeth Soria-Castro
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Edith Álvarez-León
- Subdirección de Investigación Básica y Tecnológica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Mario Nieto
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Gabriela Navarrete-Anastasio
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Wylly Ramsés García-Niño
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Stefanie Paola López-Cervantes
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana Iztapalala, Mexico City, Mexico
| | - Verónica Salas-Venegas
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Rosa Pamela Flores-Torres
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana Iztapalala, Mexico City, Mexico
| | - Armando Luna-López
- Departamento de Investigación Básica, Instituto Nacional de Geriatria, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Mina Königsberg
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico.
| |
Collapse
|
13
|
Gong L, Mai Y, Wu Z, Luo J, Wen G. Associations between iron status and diabetic kidney disease: A nationwide study. Nutr Metab Cardiovasc Dis 2025; 35:103907. [PMID: 40087039 DOI: 10.1016/j.numecd.2025.103907] [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: 08/29/2024] [Revised: 01/01/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND AND AIM Iron status plays a crucial role in various physiological processes, and its dysregulation is associated with numerous health conditions. However, research on the relationship between iron status and diabetic kidney disease (DKD) is quite limited. Therefore, this study aims to investigate the connection between iron status and DKD. METHODS AND RESULTS This population-based cross-sectional survey included adult diabetes patients from five National Health and Nutrition Examination Survey (NHANES) cycles spanning 1999 to 2006 and 2017 to 2018. Regression models were used to assess the impact of iron status on the prevalence of diabetic nephropathy. Restricted cubic spline models further explored potential nonlinear dose-response relationships. Subgroup analyses clarified the effects of other covariates on these associations. Iron and TIBC were negatively correlated with DKD, albuminuria, and low estimated glomerular filtration rate (eGFR). TSAT was negatively correlated with DKD and showed an "L"-shaped nonlinear correlation with albuminuria and low-eGFR. Ferritin exhibited a "J"-shaped nonlinear correlation with DKD, albuminuria, and low-eGFR. Subgroup analysis revealed that the association between TIBC and reduced risk of low eGFR was more pronounced in individuals with hypertension. The associations between iron and TSAT with a reduced risk of DKD were more significant in smokers, while the association between ferritin and an increased risk of albuminuria was also more pronounced in smokers. CONCLUSIONS In diabetic patients, iron status is closely associated with DKD. Monitoring these iron status markers can help improve the prevention and management of kidney health in diabetic patients.
Collapse
Affiliation(s)
- Liya Gong
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanpei Mai
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Ziqi Wu
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jingwen Luo
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ge Wen
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
14
|
Ramalho A, Vale A, Carvalho F, Fernandes E, Freitas M. Parabens exposure and its impact on diabesity: A review. Toxicology 2025; 515:154125. [PMID: 40132785 DOI: 10.1016/j.tox.2025.154125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Parabens are a family of alkyl esters of 4-hydroxybenzoic acid. The most commonly used include methylparaben, ethylparaben, propylparaben, and butylparaben. These compounds have been reported to disrupt the endocrine system and are believed to affect the central nervous, immune, and reproductive systems, as well as lipid homeostasis, glucose levels, and thyroid function. Given these effects, parabens pose potential health risks, including their possible link to conditions like diabesity - a term describing the dual condition of type 2 diabetes mellitus and obesity. This review explores current literature on how parabens may influence key mechanisms in diabesity, such as gluconeogenesis, glycogenolysis, adipogenesis, insulin resistance, and inflammation. Understanding their role in these metabolic pathways is critical for assessing their contribution to the diabesity epidemic and guiding future research for minimizing their harmful health impacts.
Collapse
Affiliation(s)
- Ana Ramalho
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Abel Vale
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Félix Carvalho
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050‑313, Portugal; Associated Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal.
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal.
| |
Collapse
|
15
|
Fang H, Sun X, Ding Y, Niu B, Chen Q. Loureirin B analogs mitigate oxidative stress and confer renal protection. Cell Signal 2025; 132:111787. [PMID: 40188928 DOI: 10.1016/j.cellsig.2025.111787] [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: 03/13/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Diabetic kidney disease (DKD) is a microvascular complication of diabetes with high morbidity and mortality, necessitating effective treatment. In this study, the Loureirin B analogue (LB-A) was utilized to treat DKD in mice. The results demonstrated that LB-A effectively prevent the progression of DKD in mice, significantly lowering fasting blood glucose levels and reducing proteinuria levels. Additionally, there was a significant decrease in oxidase content in the kidneys of mice, accompanied by an increase in antioxidant oxidase content, resulting in a decrease in ROS levels, mitigating oxidative stress state through modulation of Cxcl1. Cell experiments further confirmed that reducing Cxcl1/Cxcr2 axis activation prevented the onset of DKD induced by high glucose exposure and affected the therapeutic effect of LB-A as well. These findings provide evidences to support that LB-A may mitigate oxidative stress by modulating the Cxcl1 signaling pathway, thereby contributing to renal protection in the context of DKD treatment.
Collapse
Affiliation(s)
- Haowen Fang
- School of environmental and chemical engineering, Shanghai University, Shanghai, PR China.
| | - Xiaodong Sun
- Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Medicine, Shanghai University, Shanghai, PR China.
| | - Yanting Ding
- Tongji Hospital, School of Medicine, Tongji University, Shanghai, PR China; Shanghai Biochip Co., Ltd., National Engineering Center for Biochip at Shanghai, Shanghai, PR China.
| | - Bing Niu
- School of Life Sciences, Shanghai University, Shanghai, PR China.
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai, PR China.
| |
Collapse
|
16
|
Fu Y, Wang C, Sun W, Kong H, Liang W, Shi T, Li Q, Jia M, Zhao W, Song H. MINT3 promotes STING activation and facilitates antiviral immune responses. Cell Signal 2025; 132:111825. [PMID: 40254147 DOI: 10.1016/j.cellsig.2025.111825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Stimulator-of-interferon genes (STING) translocation is the rate-limiting step in the cGAS-STING signaling which detects cytosolic DNA and produces type I interferons. However, the mechanism by which this process is modulated remains to be further clarified. In the present study, we identified munc18-1-interacting protein 3 (MINT3) as a positive regulator of STING signaling. MINT3 promotes type I interferons production induced by herpes simplex virus-1 (HSV-1) infection and ISD or cGAMP stimulation in mouse peritoneal macrophages. Deficiency of Mint3 greatly inhibited STING and IRF3 activation in macrophages. Mint3 knockdown also attenuated STING and IRF3 activation in macrophages, human THP-1 cells, and RAW264.7 cells. Mechanistically, MINT3 interacted with STING, selectively enhanced its K63-linked polyubiquitination and facilitated STING translocation to the Golgi, resulting in the enhancement of the STING and TBK1 interaction. Furthermore, MINT3 also facilitated HSV-1-induced innate antiviral immune responses and impaired HSV-1 replication in vitro and in vivo. Interestingly, we showed that the expression of MINT3 was dramatically elevated during HSV-1 infection, and ISD stimulation in macrophages. Thus, we have revealed a feedback mechanism for the regulation of the cGAS-STING pathway, providing a promising therapeutic target for the treatment of disorders triggered by aberrant STING activity.
Collapse
Affiliation(s)
- Yue Fu
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Caiwei Wang
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenyue Sun
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hongyi Kong
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenbo Liang
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tongrui Shi
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qizhao Li
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mutian Jia
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Zhao
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Song
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
17
|
Zhang X, Hao C, Li T, Gao W, Ren Y, Wang J, Zhang Y. Leptin attenuates diabetic cardiomyopathy-induced cardiac remodeling via regulating cGAS/STING signaling and Opa1-mediated mitochondrial fusion. Cell Signal 2025; 132:111805. [PMID: 40246132 DOI: 10.1016/j.cellsig.2025.111805] [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: 03/11/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE This investigation seeks to elucidate the contribution of leptin to the pathogenesis of diabetic cardiomyopathy (DCM). METHODS Mice were rendered diabetic through the administration of streptozotocin (STZ). Leptin was delivered via subcutaneously implanted osmotic pumps. Assessments of cardiac performance, hypertrophy, and fibrosis were conducted using echocardiography, Hematoxylin and Eosin (H&E), Wheat Germ Agglutinin (WGA), and Masson trichrome staining. Myocardial apoptosis and oxidative stress were quantified through TUNEL assay and biochemical markers of oxidative stress, including Malondialdehyde (MDA), 4-Hydroxynonenal (4-HNE), and 3-Nitrotyrosine (3NT). Mitochondrial structure was examined using Transmission Electron Microscopy (TEM). Primary neonatal cardiomyocytes were subjected to high glucose (HG) conditions. The fluorescent indicators MitoTracker Green and MitoSOX Red were employed to evaluate mitochondrial morphology and function within the cardiomyocytes. RESULTS Mice with diabetes displayed marked cardiac hypertrophy and fibrosis, as indicated by H&E, WGA, and Masson staining. The administration of leptin significantly mitigated the cardiac pathological manifestations in diabetic mice. Leptin increased the expression of Opa1 and enhanced mitochondrial fusion and function in cardiomyocytes exposed to HG. The cGAS/STING signaling pathway may serve as a pivotal intermediary for leptin to facilitate Opa1-driven mitochondrial fusion. CONCLUSIONS Leptin appears to safeguard against hyperglycemia-induced mitochondrial oxidative damage and DCM by modulating the cGAS/STING signaling cascade and Opa1-mediated mitochondrial fusion. These results propose that leptin could be a promising agent for promoting mitochondrial fusion and preventing diabetes-associated cardiac pathologies.
Collapse
Affiliation(s)
| | - Chunyuan Hao
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Tonghua Li
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Weihua Gao
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Yang Ren
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Junzhe Wang
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China
| | - Yuyang Zhang
- Cardiovascular Department, Xi'an No.1 Hospital, Xi'an, Shaanxi, China.
| |
Collapse
|
18
|
Zhu Y, Yang J, Zhang JL, Liu H, Yan XJ, Ge JY, Wang FF. Dapagliflozin activates the RAP1B/NRF2/GPX4 signaling and promotes mitochondrial biogenesis to alleviate vascular endothelial ferroptosis. Cell Signal 2025; 132:111824. [PMID: 40280228 DOI: 10.1016/j.cellsig.2025.111824] [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: 01/14/2025] [Revised: 03/25/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Vascular endothelial ferroptosis is a key mechanism underlying endothelial injury and atherosclerotic plaque formation. Dapagliflozin, an essential medication in the management of heart failure, has been shown to delay atherosclerosis progression. However, the underlying mechanisms remain unclear. This study aimed to elucidate the molecular pathways whereby dapagliflozin inhibits vascular endothelial ferroptosis. We utilized human umbilical vein endothelial cells (HUVECs) to construct a cell model of atherosclerosis combined with ferroptosis. Dapagliflozin significantly decreased the iron and malondialdehyde levels and the release of inflammatory factors in HUVECs treated with oxidized low-density lipoprotein or Erastin but increased the superoxide dismutase activity and the reduced glutathione / oxidized glutathione ratio. Results from transmission electron microscopy indicated that dapagliflozin alleviated the mitochondrial shrinkage and the reduction in the number of cristae in these HUVECs. RNA sequencing revealed that dapagliflozin upregulates RAP1B. In vitro experiments showed that RAP1B upregulates NRF2 and promotes its nuclear translocation, activating the xCT/GPX4 signaling pathway and inhibiting lipid peroxidation. Additionally, dapagliflozin induces mitochondrial biogenesis and enhances oxidative phosphorylation through the RAP1B/NRF2 pathway, reducing iron overload and excessive production of mitochondrial reactive oxygen species, ultimately mitigating ferroptosis. At the animal level, we constructed an atherosclerosis model by using Apoe-/-; Rap1b-/- double-knockout mice. Rap1b knockout blocked the inhibitory effects of dapagliflozin on atherosclerotic plaque formation and ferroptosis activation. We confirmed in vivo that dapagliflozin upregulates GPX4 and key factors of mitochondrial biogenesis via RAP1B, promoting oxidative phosphorylation. When mitochondrial oxidative phosphorylation was pharmacologically inhibited, ferroptosis was reactivated, promoting atherosclerotic plaque formation. In conclusion, this study demonstrated that dapagliflozin activates the RAP1B/NRF2/GPX4 signaling pathway and promotes mitochondrial biogenesis, thereby alleviating vascular endothelial ferroptosis.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, PR China
| | - Jin Yang
- Pfizer, Inc., Cambridge, MA, USA
| | - Jia-Li Zhang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213000, PR China
| | - Hao Liu
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, PR China
| | - Xue-Jiao Yan
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, PR China
| | - Ji-Yong Ge
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, PR China
| | - Fang-Fang Wang
- Department of Cardiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, PR China.
| |
Collapse
|
19
|
Wang Q, Liu H, Yuan M, Wang Y, Qin J, Chen X, Lei Z, Song X, Wu X. OTUD1 inhibits macrophage ferroptosis via regulation of AMPK and GSK3β/β-catenin signaling pathways exerting protective effects in sepsis-induced acute lung injury. Int Immunopharmacol 2025; 160:114985. [PMID: 40460602 DOI: 10.1016/j.intimp.2025.114985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/24/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025]
Abstract
Sepsis is a life-threatening organ dysfunction caused by dysregulated inflammatory and immune responses to infection. Its global incidence and mortality remain high, posing a severe threat to public health. Acute lung injury (ALI) is a common and serious complication of sepsis. Current understanding of the pathogenesis and effective therapeutic strategies for sepsis-induced acute lung injury (SI-ALI) remains insufficient. This study aims to investigate the role and underlying mechanisms of the deubiquitinase OTUD1 in sepsis-induced pulmonary injury. Using a mouse model of sepsis-induced lung injury combined with genetic knockout techniques and ferroptosis inhibitors, we systematically analyzed the protective effects of OTUD1 in sepsis-related lung damage and explored the regulatory roles of AMPK and GSK3β/β-catenin signaling pathways. Results demonstrated that OTUD1 gene deletion exacerbated lung tissue damage and inflammatory responses in septic mice while increasing ferroptosis levels; pretreatment with the ferroptosis inhibitor Ferrostatin-1 significantly ameliorated these effects. Further mechanistic studies revealed that OTUD1 may regulate ferroptosis levels in lung tissue by modulating the activation status of AMPK and GSK3β/β-catenin pathways. Specifically, OTUD1 may remove K63-linked ubiquitin chains from AMPK, altering its protein conformation and subsequently promoting AMPK phosphorylation to regulate the GSK3β/β-catenin signaling cascade. Collectively, this study provides the first systematic elucidation of OTUD1's protective role in sepsis-induced lung injury and its relationship with ferroptosis, offering novel molecular targets and theoretical foundations for the treatment of sepsis-associated pulmonary damage.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Huifan Liu
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yuxuan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jingxue Qin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xue Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zihan Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xuemin Song
- Department of Anesthesiology, Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
20
|
Niu ZC, Jin Q, Shen QY, Shi HH, Shang Y, Guo XT, Wang M, Yuan Q, Mao HP. Schisandrol B alleviated diabetic cardiac injury by inhibiting ferroptosis and improving lipid metabolism in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156902. [PMID: 40450978 DOI: 10.1016/j.phymed.2025.156902] [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: 02/12/2025] [Revised: 04/21/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a major complication of diabetes mellitus, highlighting the need to elucidate its pathogenesis and explore potential therapeutic interventions. PURPOSE This study aimed to investigate the cardioprotective mechanisms of SolB in DCM using metabolomic and transcriptomic approaches. METHODS A DCM mouse model was induced by a high-fat diet combined with streptozotocin (STZ) administration. Cardiac function was assessed, and myocardial structure was examined via echocardiography and HE staining after 10 weeks of SolB treatment. Serum metabolomics and cardiac transcriptomics were performed to identify differentially expressed metabolites and genes, respectively, followed by correlation analysis. Ferroptosis-related proteins were detected by Western blotting (WB). In vitro, H9c2 cells exposed to palmitic acid and high glucose were used to evaluate the effects of SolB on cell viability, ATP production, oxygen consumption, reactive oxygen species (ROS) levels, and mitochondrial membrane potential. Ferroptosis inducer and inhibitor were employed to further explore the underlying mechanisms. RESULTS SolB did not significantly alter blood glucose levels but markedly improved cardiac function and myocardial structure. Metabolomic analysis revealed that SolB modulated serum metabolic pathways, including carnitine synthesis and fatty acid oxidation et al. Transcriptomic data indicated that SolB influenced ferroptosis-related pathways. Integrated analysis demonstrated that SolB regulated fatty acid degradation, glutathione metabolism, and cysteine and methionine catabolism. In H9c2 cells, SolB enhanced cell viability, suppressed ferroptosis, reduced lactate dehydrogenase (LDH) release, and improved mitochondrial function. CONCLUSIONS SolB ameliorates diabetic myocardial injury by inhibiting ferroptosis and improving myocardial lipid metabolism.
Collapse
Affiliation(s)
- Zi-Chang Niu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Qi Jin
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qian-You Shen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hui-Hui Shi
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ye Shang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiao-Tong Guo
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Meng Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qin Yuan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Hao-Ping Mao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China, 301617; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
21
|
Li L, Huang K, Wang D, Yang D, Gao K, Tang J, Wei X, Pu H, Yu J, Zhang D, Guo S. Chlorogenic acid and ferulic acid in SMYAD alleviate diabetic cardiomyopathy by inhibiting cardiac lipotoxicity via GCGR/PPARα and GCGR/AMPK pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156906. [PMID: 40466507 DOI: 10.1016/j.phymed.2025.156906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/11/2025] [Accepted: 05/24/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) causes a high risk of heart failure, necessitating effective therapies. The Si-Miao-Yong-An decoction (SMYAD) has been widely applied in the clinical management of diabetes mellitus and cardiovascular diseases within the paradigm of traditional Chinese medicine (TCM), but its active constituents and mechanism of the action against DCM are poorly understood. PURPOSE The aim of this study was to evaluate the effects of chlorogenic acid (CGA) and ferulic acid (FA), the main active ingredients of SMYAD, on cardiac function and cardiac lipotoxicity in mice with DCM, as well as to investigate their potential molecular mechanisms. METHODS The cardioprotective effects of CGA and FA on DCM mice were identified by echocardiography, HE, Masson, and Oil Red O. Immunofluorescence, flow cytometry, MitoTracker Green staining, and seahorse analysis were used to study the effects of CGA and FA on palmitic acid (PA)-induced lipotoxicity in cardiomyocytes. Finally, the regulatory effects of CGA and FA on GCGR/PPARα and GCGR/AMPK signalling pathways were detected by Ad GCGR-infection, molecular docking, RT-PCR and western blot. RESULTS Based on previously identified 20 representative blood prototype components and in vitro multilayer lipid toxicity analysis, chlorogenic acid (CGA) and ferulic acid (FA) were screened as the main active components of SMYAD against DCM. In vivo experiments showed that CGA and FA attenuated lipotoxicity associated with cardiac GLC/GCGR in DCM mice, thereby protecting cardiac function. In vitro results showed that GCGR inhibitor (Adomeglivant) reduces PA-induced apoptosis, indicating that PA leads to cardiomyocyte lipotoxic apoptosis by activating GCGR. Moreover, CGA and FA inhibit PA-induced cardiomyocyte lipotoxic apoptosis, mitochondrial dysfunction, and energy substrate transition through inhibiting GCGR/PPARα and GCGR/AMPK pathways. Furthermore, GLC-stimulated/GCGR-infected H9c2 cardiomyocyte lipotoxic apoptosis and downstream proteins were effectively suppressed by CGA and FA, which is consistent with the effect of Adomeglivant. Docking results showed that ASP1018 and THR1024 of GCGR are the principal molecular targets for both CGA and FA. CONCLUSION GLC lipotoxic signaling is a crucial target in mediating cardiac lipotoxicity development. CGA and FA could inhibit DCM by regulating the GCGR/PPARα and GCGR/AMPK pathway, offering a novel strategy for the development of anti-DCM drugs.
Collapse
Affiliation(s)
- Lin Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing 100010, China
| | - Kai Huang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Deshuang Yang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiayang Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoqi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haiyin Pu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiang Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dongwei Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
22
|
Ko JH, Jung TW, Pyo MK, Lim DS, Gwon HJ, Abd El-Aty AM, Yağan R, Shin JW, Lee MY, Song JH, Jeong JH. Senkyunolide A mitigates lipid deposition in hyperlipidemic hepatocytes through SIRT6-mediated suppression of oxidative stress and fetuin-A. Biochem Biophys Res Commun 2025; 771:152046. [PMID: 40403679 DOI: 10.1016/j.bbrc.2025.152046] [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: 05/14/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
Senkyunolide A (SenA), a phthalide compound isolated from celery seed essential oil, is known for its anti-inflammatory and antioxidant properties in various disease models. However, its impact on hepatic lipid metabolism and associated molecular mechanisms remain unclear. This study investigated the protective effects of SenA against lipotoxicity-induced hepatic steatosis and explored its underlying pathways. Lipid accumulation was assessed via Oil Red O staining, whereas protein expression was analyzed via Western blotting. Apoptosis was evaluated through TUNEL staining, caspase-3 activity, and cell viability assays. Oxidative stress was measured via DCFDA-based ROS detection and assays for MDA and H2O2. The role of SIRT6 was examined through siRNA-mediated knockdown. SenA treatment significantly reduced lipid deposition, apoptosis, oxidative stress, and fetuin-A expression in palmitate-treated hepatocytes. It also increased the expression of SIRT6 and Nrf2, two key regulators of metabolic and redox homeostasis. Silencing SIRT6 diminished these protective effects, indicating its essential role in mediating SenA activity. These findings suggest that SenA mitigates hepatic steatosis by modulating oxidative stress and metabolic dysfunction via SIRT6/Nrf2 signaling. As a natural bioactive compound, SenA offers promise for the development of safer therapeutic strategies for managing nonalcoholic fatty liver disease (NAFLD), which has broader relevance to metabolic health.
Collapse
Affiliation(s)
- Jun Hwi Ko
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Min Kyung Pyo
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Do Su Lim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hyeon Ji Gwon
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey.
| | - Rıdvan Yağan
- Dr. Filiz Dolunay Family Health Center Unit Number:61 Erzurum, Turkey
| | - Jong Wook Shin
- Department of Internal Medicine, Division of Allergy and Respiratory Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Moo Yeol Lee
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jin-Ho Song
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Ichi MS, Shabkhiz F, Kordi M. Effects of High-Intensity Interval Training (HIIT) on miR-29c and miR-146a expression in the hippocampus of streptozotocin-induced diabetic rats. Behav Brain Res 2025; 489:115632. [PMID: 40339812 DOI: 10.1016/j.bbr.2025.115632] [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: 01/26/2025] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND MicroRNAs like miR-146a and miR-29c are potential biomarkers for diabetes, which is linked to brain impairments such as cognitive decline and hippocampal dysfunction due to hyperglycemia and inflammation. This study investigates the effects of high-intensity interval training (HIIT) on hippocampal miR-146a and miR-29c expression and serum TNF-α levels in diabetic rats, highlighting its role in reducing inflammation and improving brain function. METHODS Twenty-four male Wistar rats were divided into four groups: Control (Normal), 1-week diabetes (Diabetes 1 W), 6-week diabetes (Diabetes 6 W), and diabetic HIIT (Diabetes-Exe). Diabetes was induced using streptozotocin (55 mg/kg) and rats with blood glucose > 250 mg/dL were included. HIIT was conducted for six weeks, and hippocampal miR-146a, miR-29c expression, and TNF-α serum levels were assessed using Real-Time PCR and ELISA. TNF-α serum levels were measured as a marker of systemic inflammation. RESULTS Diabetic rats exhibited decreased miR-146a and increased miR-29c expression in the hippocampus compared to controls. Additionally, TNF-α serum levels were significantly higher in the diabetic groups, indicating an elevated inflammatory state. HIIT in the Diabetes-Exe group resulted in a non-significant change in miR-29c expression and TNF-α serum levels, accompanied by a significant increase in miR-146a expression compared to the Diabetes 6 W group. CONCLUSION HIIT exercise may help reduce hippocampal neuronal damage in diabetic rats by modulating miR-146a expression, improving blood glucose control, and reducing inflammation. Although HIIT did not significantly alter miR-29c expression, its potential as an effective non-pharmacological strategy for managing diabetic neuropathy complications cannot be excluded.
Collapse
Affiliation(s)
- Mehdi Soltani Ichi
- Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, University of tehran, Tehran, Iran.
| | - Fatemeh Shabkhiz
- Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, University of tehran, Tehran, Iran.
| | - Mohammadreza Kordi
- Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, University of tehran, Tehran, Iran.
| |
Collapse
|
24
|
Franconi F, Lodde V, Capobianco G, Criscione M, Montella A, Campesi I. Effects of maternal smoking on inflammation, autophagy/mitophagy, and miRNAs in endothelial cells: Influence of newborn sex. Eur J Pharmacol 2025; 998:177648. [PMID: 40252896 DOI: 10.1016/j.ejphar.2025.177648] [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: 01/07/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Maternal smoking (MS) during pregnancy is linked to well-documented adverse health effects for the mother and foetus, however the role of fetal sex was largely overlooked. Primary cultures of male and female human umbilical vein endothelial cells (MHUVECs and FHUVECs, respectively) were used. IL-6, IL-8, and TNF-α levels were measured in HUVECs supernatant. The expression of genes and proteins (oestrogen receptors (ERs), Hsp90, Beclin-1, p62, LC3, LAMP-1 and Parkin), as well as the expression of miR-29a-3p, miR-29b-3p, miR-126-3p, miR-133a-3p, and miR-146a-5p were analysed in cells obtained from foetuses born to non-smoking and smoking mothers. In HUVECs from foetuses born to non-smoking mothers, Beclin-1 protein was higher in MHUVECs (1.8 fold increase), while Parkin, Hsp90 proteins, and miR-146a-5p were elevated in FHUVECs (2.2, 2.6, and 2.1 fold increase, respectively), with no other significant differences. MS amplified these sex differences, with specific effects based on foetus sex. FHUVECs obtained from foetus born to smoking mothers showed higher levels of IL-8 (1399.36 ± 123.96 pg/ml for FHUVECs vs 655.11 ± 215.94; pg/ml for MHUVECs; P < 0.001), Hsp90 gene and protein (3.3 and 2.6 fold increase), and ERβ protein and Beclin-1 gene (2.1, and 4.9 fold increase), and lower levels of miR-29b-3p, miR-133a-3p, and miR-146a-5p than MHUVECs (0.27, 0.68, and 0.1 fold change, respectively). This study shows that primary HUVECs from fetuses born to smoking mothers retain a memory of smoking effects, with sex differences in gene expression, miRNA profiles, and autophagic responses, suggesting that maternal smoking impacts endothelial cell physiology in a sex-dependent manner.
Collapse
Affiliation(s)
- Flavia Franconi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100, Sassari, Italy
| | - Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy; Gynecologic and Obstetric Clinic, AOU, Viale San Pietro 12, 07100, Sassari, Italy
| | - Massimo Criscione
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100, Sassari, Italy; Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy.
| |
Collapse
|
25
|
Wang H, Peng LJ, Lu W, Li GR, Zhao PT, Lv X, Dong MQ, Liu ML. Acacetin reverses hypoxic pulmonary hypertension by inhibiting hypoxia-induced proliferation of pulmonary artery smooth muscle cells via SIRT1-HMGB1 pathway. Eur J Pharmacol 2025; 998:177650. [PMID: 40258398 DOI: 10.1016/j.ejphar.2025.177650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary arterial pressure and vascular remodeling. The present study is to investigate the efficacy of acacetin on HPH and its potential molecular mechanism. C57/BL6 mice were exposed to hypobaric hypoxia for six weeks. At 4th week of hypoxia exposure, mice were administrated with the water-soluble prodrug of acacetin (5, 10, 20 mg/kg) or equivalent normal saline for another two weeks. The haemodynamic and pathohistological assessment were performed. Primary pulmonary artery smooth muscle cells (PASMCs) were cultured to examine the anti-proliferation efficacy of acacetin (0.3-3 μM). The activity and expression of sirtuin1 (SIRT1) acetylation and distribution of high-mobility group box 1 (HMGB1) were determined in lungs and/or cultured PASMCs with or without RNA interference of SIRT1. Macromolecular docking and molecular dynamics simulation were done to explore the potential binding between acacetin and SIRT1. Results showed that acacetin prodrug significantly reversed the increased pulmonary pressure and vascular remodeling in HPH mice, which is associated with inhibiting the reduction in SIRT1 and the increase in HMGB1, and inhibiting the nucleocytoplasmic translocation of HMGB1. In cultured PASMCs, acacetin inhibited the hyper-proliferation induced by hypoxia, reversed the SIRT1 reduction and inhibited the nucleocytoplasmic translocation of HMGB1 and HMGB1 increase. Silencing SIRT1 abolished all the beneficial effects of acacetin. These results demonstrate that acacetin is very effective in reversing HPH by inhibiting PASMC hyper-proliferation via regulating SIRT1-HMGB1 signaling, suggesting that acacetin is likely a promising drug candidate for treating patients with HPH.
Collapse
Affiliation(s)
- Hui Wang
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611137, China
| | - Li-Jing Peng
- Department of Cardiology, 986th Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710054, China
| | - Wang Lu
- Endoscopic Center, Xi'an Chest Hospital, Xi'an, Shaanxi, 710100, China
| | - Gui-Rong Li
- Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu, 210032, China
| | - Peng-Tao Zhao
- School of Medicine, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Xing Lv
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Ming-Qing Dong
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, 325000, China.
| | - Man-Ling Liu
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
26
|
Xu H, Mao X, Mo D, Lv M. 6PPD impairs growth performance by inducing intestinal oxidative stress and ferroptosis in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2025; 293:110161. [PMID: 39988222 DOI: 10.1016/j.cbpc.2025.110161] [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: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD), a tire-derived pollutant, has gained increasing attention due to its potential toxicity to aquatic organisms. Although previous studies have revealed that 6PPD impacts early developmental stages of larval fish, its effects on adult fish, particularly on key organs, remain unclear. In this study, we observed that adult zebrafish exposed to 6PPD exhibited reduced growth performance and increased fecal output. Histological examination with hematoxylin and eosin (H&E) staining revealed damage to the intestinal villi and a reduction in goblet cell numbers, indicating that 6PPD impairs growth performance by disrupting the digestive system. Comparative transcriptomic analysis revealed that 6PPD caused significant changes in the expression of 727 genes in the intestine, of which 280 genes were up-regulated and 447 genes were down-regulated. These genes were primarily associated with nutrient digestion and absorption, energy metabolism, immune response, and redox regulation. Mechanistically, 6PPD induced oxidative stress and triggered ferroptosis in the intestine, leading to structural damage of the intestinal villi. Treatment with the antioxidant N-acetylcysteine (NAC) alleviated 6PPD-induced oxidative stress and ferroptosis, thereby improving intestinal villi structure and promoting fish growth. This study provides insights into the mechanisms by which 6PPD impairs growth in adult zebrafish and highlights NAC as a potential therapeutic strategy to mitigate its toxicity.
Collapse
Affiliation(s)
- Hao Xu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Aquaculture Engineering Technology Research Center, College of Fisheries, Southwest University, Chongqing 400715, China.
| | - Xiaoyu Mao
- College of Language Intelligence, Sichuan International Studies University, Chongqing 400031, China
| | - Dashuang Mo
- Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| | - Mengzhu Lv
- Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China.
| |
Collapse
|
27
|
Bai H, Meng F, Ke K, Fang L, Xu W, Huang H, Liang X, Li W, Zeng F, Chen C. The significance of small noncoding RNAs in the pathogenesis of cardiovascular diseases. Genes Dis 2025; 12:101342. [PMID: 40247912 PMCID: PMC12005926 DOI: 10.1016/j.gendis.2024.101342] [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: 12/06/2023] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 04/19/2025] Open
Abstract
With the advancement of high-throughput sequencing and bioinformatics, an increasing number of overlooked small noncoding RNAs (sncRNAs) have emerged. These sncRNAs predominantly comprise transfer RNA-derived fragments (tsRNAs), PIWI-interacting RNAs (piRNAs), Ro-associated non-coding RNAs (RNYs or Y-RNAs), small nucleolar RNAs (snoRNAs), and small nuclear RNAs (snRNAs). Each of these RNA types possesses distinct biological properties and plays specific roles in both physiological and pathological processes. The differential expression of sncRNAs substantially affects the occurrence and progression of various systemic diseases. However, their roles in the cardiovascular system remain unclear. Therefore, understanding the functionality and mechanisms of sncRNAs in the cardiovascular system holds promise for identifying novel targets and strategies for the diagnosis, prevention, and treatment of cardiovascular diseases. This review examines the biological characteristics of sncRNAs and their potential roles in cardiovascular diseases.
Collapse
Affiliation(s)
- Hemanyun Bai
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Fanji Meng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Kangling Ke
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Lingyan Fang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Weize Xu
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Haitao Huang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Xiao Liang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Weiyan Li
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Fengya Zeng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Can Chen
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| |
Collapse
|
28
|
Kirkutyte I, Latunde-Dada GO. Relationship Between Hepatic Iron Concentration and Glycemic Metabolism, Prediabetes, and Type 2 Diabetes: A Systematic Review. Nutr Rev 2025; 83:e1828-e1842. [PMID: 39724915 DOI: 10.1093/nutrit/nuae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
CONTEXT Emerging research has suggested a potential link between high iron levels, indicated by serum ferritin levels, and the development of type 2 diabetes (T2D). However, the role of hepatic iron concentration (HIC) on T2D development and progression is not well understood. OBJECTIVES This study aims to systematically review the literature on HIC and/or the degree of hepatic iron overload (HIO) in individuals with prediabetes and/or diagnosed T2D, and to analyze associations between HIC and markers of glucose metabolism. DATA SOURCES The databases Medline, PubMed, Embase, CINAHL, and Web of Knowledge were searched for studies published in English from 1999 to March 2024. This review followed the Preferred Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist. DATA EXTRACTION Data were extracted following the established eligibility criteria. Study characteristics and biomarkers related to prediabetes, T2D, and HIO were extracted. The risk of bias was analyzed using the Newcastle-Ottawa Scale. Data were stratified by the exposure and analyzed in subgroups according to the outcome. Data regarding the HIC values in controls, individuals with prediabetes, and individuals with T2D and the association estimates between HIC or HIO and markers of glycemic metabolism, prediabetes, or T2D were extracted. DATA ANALYSIS A total of 12 studies were identified, and data from 4110 individuals were analyzed. HIO was not consistently observed in prediabetic/T2D populations; however, elevated HIC was frequently observed in prediabetic and T2D individuals, and was associated with the disruption of certain glycemic markers in some cases. CONCLUSION The extent of iron overload, as indicated by hepatic iron load, varied among the prediabetic and T2D populations studied. Further research is needed to understand the distribution and regulation of iron in T2D pathology.
Collapse
Affiliation(s)
- Indre Kirkutyte
- Department of Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, United Kingdom
| | - Gladys Oluyemisi Latunde-Dada
- Department of Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, United Kingdom
| |
Collapse
|
29
|
Tang YX, Cao HL, Hou JR, Chen J, Zhu SY, Liang YS, Yang ZY, Malhi KK, Mao XY, Li JL. A new insight into the mechanism of cadmium-induced nephrotoxicity and antagonism of selenium nanoparticles: modulation of MTF1-MT axis dependent metal homeostasis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126320. [PMID: 40324627 DOI: 10.1016/j.envpol.2025.126320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/06/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Cadmium (Cd) is a highly toxic pollutant that causes direct harm to the kidney. Cellular metal homeostasis is one of the key targets of heavy metal toxicity and may represent an alleviation pathway for environmentally-related kidney disease. Selenium nanoparticles (SeNPs) is a antioxidant and is considered to be involved in the regulation of cellular metal transport. However, the impact of Cd on renal metal homeostasis and the protective role of SeNPs against Cd-induced nephrotoxicity have not been fully elucidated. Chickens were treated with CdCl2 and SeNPs during this 90-day trial. SeNPs alleviated Cd-induced kidney injury, preserved filter barrier, and restored disrupted metal transport and selenoprotein biosynthesis, thereby normalizing the renal Cd, Se, Zn, Fe, and Cu levels. Further investigation identified that SeNPs increased nuclear translocation of MTF1 and the transcription of downstream genes MT1 and MT2. JNK activated MTF1 under a low dose of Cd exposure in response to metal stress. However, high doses of Cd exposure led to MTF1 dysfunction and Cd accumulation. Interestingly, SeNPs upregulated MTF1 expression despite down-regulation of JNK phosphorylation, suggesting that SeNPs activated the MTF1-MT axis in a non-JNK-dependent pathway. These results support the key role of SeNPs in responding to metal stress.
Collapse
Affiliation(s)
- Yi-Xi Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Hui-Lin Cao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jia-Rong Hou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jian Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yun-Shuang Liang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhou-Yi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Kanwar Kumar Malhi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xin-Yue Mao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
30
|
Zierold KM, Smith MJ, Xu J, Cai L, Sears L. Distinct Features of Proton-Induced X-ray Emission (PIXE) and Inductively Coupled Plasma Mass Spectrometry (ICP-MS) When Used for Elemental Analysis of Nail Samples. Cardiovasc Toxicol 2025; 25:969-978. [PMID: 40377879 PMCID: PMC12159087 DOI: 10.1007/s12012-025-10011-9] [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: 12/20/2024] [Accepted: 04/29/2025] [Indexed: 05/18/2025]
Abstract
Biomarkers, such as toenails, are commonly used to investigate the health status of individuals. Nails samples are a useful marker of exposure, as they are easy to collect, store, and represent exposure from 6 to 12 months. There are multiple analytical methods that can be used to extract long-term exposure profiles from toenails including Proton-Induced X-Ray Emission(PIXE) and Inductively Coupled Plasma Mass Spectrometry (ICP-MS). The overall goal of this research was to evaluate the level of agreement between the two analytical methods for assessment of different metals in nail samples. Children's nail samples were collected. Nail samples were first analyzed by PIXE and then analyzed by ICP-MS. To compute quantiles for the metal concentrations that had some fully observed and some left-censored concentrations, a reverse Kaplan-Meier estimator was used. Lin's concordance correlation coefficient (CCC) and the Pearson correlation coefficient were calculated to assess agreement between the two methods and to determine the strength of the linear association between the metal concentration measurements obtained under each analytical technique. PIXE and ICP-MS determined similar median concentrations for calcium, copper, potassium, and nickel. However, there were stark differences between other elements. Several elements, such as copper, potassium, and zinc represented strong concordance through use of the CCC. In many studies, scholars want to evaluate how well one measurement can reproduce another, and our paper used several elements to show the degree of reproducibility between the two analytical methods. This can be useful when scholars are determining methods to assess biomarkers in health-related studies.
Collapse
Affiliation(s)
- Kristina M Zierold
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, RPHB 534 C, 1720 2nd Ave S, Birmingham, AL, 35294-0022, USA.
| | - Melissa J Smith
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason Xu
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY, USA
- Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lonnie Sears
- The Center for Integrative Environmental Health Sciences, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
31
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
32
|
Yin J, Song Z, Zhang L, Cong J. Methylophiopogonanone A alleviates diabetic cardiomyopathy via inhibiting JNK1 signaling. Cell Signal 2025; 131:111762. [PMID: 40139620 DOI: 10.1016/j.cellsig.2025.111762] [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: 03/23/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Diabetic cardiomyopathy (DCM) is a common complication of type 2 diabetes mellitus (T2DM). The effects of methylophiopogonanone A (MO-A), a natural homoisoflavonoid with anti-inflammatory effects, on DCM and its underlying mechanisms were investigated in this study. METHODS The T2DM mouse model was induced by intraperitoneal injection of 30 mg/kg streptozotocin for 7 consecutive days and fed with a high-fat diet for 12 weeks. T2DM mice received MO-A (2.5, 5, or 10 mg/kg) treatment for two weeks. Cardiac function, hypertrophy, fibrosis, and inflammation were evaluated. The binding energy between MO-A and JNK1 was analyzed using molecular docking. The underlying mechanism was further investigated in high glucose (HG)-induced H9C2 cells. The cytotoxic effects, cardiomyocyte hypertrophy, fibrosis, inflammation, and relevant signaling proteins were assessed. RESULTS MO-A treatment alleviated cardiac function and histopathological changes in DCM mice. Moreover, MO-A treatment significantly decreased COLI, TGF-β1, MYH7, and ANP expression levels in DCM mice. Furthermore, TNF-α, IL-6, and IL-1β expression levels were notably downregulated after treatment with MO-A in DCM mice. Similar results were also observed in vitro. Mechanistically, MO-A targets JNK1 and downregulates its phosphorylation levels in DCM mice. The protective properties of MO-A were reversed by JNK1 overexpression in HG-induced H9C2 cells. CONCLUSION Our results revealed that MO-A could alleviate cardiac function, hypertrophy, fibrosis, and inflammation in DCM via inhibiting JNK1 signaling.
Collapse
Affiliation(s)
- Jing Yin
- Department of Traditional Chinese Medicine, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Zhicheng Song
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong, China
| | - Lijun Zhang
- Department of Endocrinology, Longkou Traditional Chinese Medicine Hospital, Yantai 265701, Shandong, China
| | - Jialin Cong
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong, China.
| |
Collapse
|
33
|
Lin Y, Xu T, Jiang Q, Chen J, Zhang H, Reinach PS, Yan D, Qu J, Chen S. Fluorometholone inhibits corneal epithelial proliferation, migration via targeting Rho GTPases: RhoA, Rac1, and Cdc42. Exp Eye Res 2025; 256:110397. [PMID: 40268157 DOI: 10.1016/j.exer.2025.110397] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Abnormal corneal epithelial hyperplasia is a common complication following refractive surgery. 0.1 % fluorometholone (FML) eye drops are commonly used for treatment. However, their efficacy varies among patients, potentially attributed to differences in the patient's microenvironment. The underlying reason remains incompletely understood. This study aimed to elucidate the molecular mechanisms of FML's action on corneal epithelial cells (CECs). The effects of FML on the cell viability, proliferation, cell cycle, and migration of human corneal epithelial cells (HCECs) were evaluated using MTS assay, EdU staining, flow cytometry, and scratch assay, respectively. Mouse corneal sections were immunofluorescently stained to assess cell proliferation. A corneal wound model, monitored by slit-lamp photography, was utilized to evaluate the impact of FML on wound healing. Gene expression alterations were detected via RNA sequencing. RT-qPCR and Western blot were employed to validate gene and protein expression in HCECs and mouse corneal epithelia. Proteomic analysis was conducted on tear samples from patients. FML treatment significantly inhibited CEC proliferation, migration, and wound healing. At the molecular level, FML treatment led to a remarkable downregulation of RhoA, Rac1, and Cdc42. Correspondingly, reductions in the downstream Erk and NF-κB signaling pathways were observed in both HCECs and mouse corneal epithelia. Moreover, these pathways were similarly downregulated in tear samples from clinical patients. In conclusion, FML inhibits CEC proliferation and migration by modulating the Rho GTPase signaling network, especially through RhoA/Rac1/Cdc42, thereby suppressing the Erk/NF-κB pathway.
Collapse
Affiliation(s)
- Yong Lin
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tianyi Xu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiuruo Jiang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jialu Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hua Zhang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peter Sol Reinach
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Yan
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Shihao Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
34
|
Hashemi M, Gholamrezaie H, Ziyaei F, Asadi S, Naeini ZY, Salimian N, Enayat G, Sharifi N, Aliahmadi M, Rezaie YS, Khoushab S, Rahimzadeh P, Miri H, Abedi M, Farahani N, Taheriazam A, Nabavi N, Entezari M. Role of lncRNA PVT1 in the progression of urological cancers: Novel insights into signaling pathways and clinical opportunities. Cell Signal 2025; 131:111736. [PMID: 40081549 DOI: 10.1016/j.cellsig.2025.111736] [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: 07/31/2024] [Revised: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Urologic malignancies, encompassing cancers of the kidney, bladder, and prostate, represent approximately 25 % of all cancer cases. Recent advances have enhanced our understanding of PVT1's crucial functions. Long noncoding RNAs influence both the onset and development of cancer, as well as epigenetic alterations. Recent findings have focused on PVT1's mechanism of action across several malignancies, particularly urologic cancers. Understanding the various functions of PVT1 linked to cancer is necessary for the development of cancer detection and treatment when PVT1 is dysregulated. Furthermore, recent advancements in genomic and epigenetic research have elucidated the complex regulatory networks that control PVT1 expression. Comprehending the intricate role of PVT1 Understanding the complex function of PVT1 in urologic cancers has substantial clinical implications. Here, we summarize some of the most recent findings about the carcinogenic effects of PVT1 signaling pathways and the possible treatment strategies for urological malignancies that target these pathways.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Gholamrezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Ziyaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Yousefian Naeini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology,Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Salimian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Golnaz Enayat
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nafiseh Sharifi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Melika Aliahmadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Soofi Rezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran,Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
35
|
Li H, Wang G, Tang Y, Wang L, Jiang Z, Liu J. Rhein alleviates diabetic cardiomyopathy by inhibiting mitochondrial dynamics disorder, apoptosis and hypertrophy in cardiomyocytes. Cell Signal 2025; 131:111734. [PMID: 40081546 DOI: 10.1016/j.cellsig.2025.111734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/18/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a significant cardiovascular complication in diabetic patients, and treatment regimens are limited. Rhein, a compound extracted from the herb rhubarb, was investigated in this study for its efficacy on DCM and the potential mechanism. METHODS Streptozotocin-induced DCM mice, high-glucose (HG)-treated neonatal rat cardiomyocytes (NRCMs), and H9c2 cells with ClpP knockdown were used for the study. We performed phenotypic and molecular mechanistic studies using immunoblotting, quantitative polymerase chain reaction, transmission electron microscopy, cardiac echocardiography, and histopathological analysis. RESULTS Rhein improved the cardiac function and myocardial fibrosis, and decreased the cross-sectional area of cardiomyocytes in the DCM mice. It also improved mitochondrial dynamic disorder as evidenced by a decreased ratio of mitochondrial fission-related proteins p-Drp1S616/ Drp1 and increased expression of mitochondrial fusion proteins (Opa1, Mfn1 and Mfn2). Rhein mitigated apoptosis as indicated by decreased apoptosis-related proteins (caspase 9, cleaved-caspase 3 and Bax) and increased anti-apoptosis protein Bcl2 in the heart tissue of DCM mice. Upregulations of cardiac hypertrophy associated genes (ANP, BNP and β-MHC) were significantly inhibited by Rhein treatment. In addition, the level of ClpP, a mitochondrial protease, was increased in DCM, but was normalized by Rhein treatment. However, ClpP knockdown exacerbated cardiomyocyte injury in the presence or absence of HG in H9c2 cells, indicating that a normal level of ClpP is essential for cardiomyocytes to survive. CONCLUSIONS Our results suggest that Rhein protects DCM by ameliorating mitochondrial dynamics disorder, inhibiting cardiomyocyte apoptosis, and myocardial hypertrophy. These protective effects of Rhein may be mediated by preventing ClpP upregulation.
Collapse
Affiliation(s)
- Hejuan Li
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Genwang Wang
- Department of Health Service, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yi Tang
- Department of Cardiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lei Wang
- Department of Cardiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China.
| | - Jing Liu
- Department of Cardiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
36
|
Kang P, Xiao L, Liu Y, Yang J, Li S, Wang L. Morusin ameliorates tubulointerstitial damage in diabetic mice through SIRT1/HIF-1α/IL-16 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156781. [PMID: 40382816 DOI: 10.1016/j.phymed.2025.156781] [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: 09/26/2024] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND PURPOSE Hypoxia is generally considered the major cause of renal tubular injury. The interaction between sirtuin 1 (SIRT1) and hypoxia inducible factor 1 subunit alpha (HIF-1α) is a key mediator of hypoxia-induced renal tubular damage. In this study, we identified morusin from a screen of SIRT1 inducers and reported its potential for the treatment of diabetic kidney disease (DKD). EXPERIMENTAL APPROACH Intraperitoneal injection of morusin in db/db and STZ-induced diabetic mice was assessed for its effect on tubulointerstitial damage. The expression and acetylation level of HIF-1α were analyzed by immunoblotting or immunostaining. The effects of morusin treatment on hypoxia-induced extracellular matrix (ECM) accumulation, apoptosis and IL-16 production in HK2 cells were evaluated by immunoblotting. Luciferase reporter gene and ChIP analysis was used to determine whether IL-16 was the target genes of HIF-1α. KEY RESULTS In db/db and STZ-induced mice, morusin treatment alleviated kidney injury and inhibited renal inflammation and fibrosis. Mechanistic analysis using animal models and HK2 cells revealed that morusin treatment upregulated SIRT1 expression by enhancing its stability, and reduced the expression and acetylation levels of HIF-1α as well as expression of its regulated genes, thereby inhibiting ECM accumulation, apoptosis and IL-16 production. Furthermore, morusin decreased expression of the IL-16 through inhibited HIF-1α binding to the IL-16 promoter. CONCLUSION AND IMPLICATIONS Our study is the first to demonstrate that morusin ameliorated tubulointerstitial damage in diabetic mice by regulating SIRT1/HIF-1α/IL-16 signaling pathway. Taken together, our findings suggest that morusin is a clinical candidate compound to prevent DKD.
Collapse
Affiliation(s)
- PeiYuan Kang
- Department of Clinical Medicine, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Lin Xiao
- Department Of Physiology, Hebei Medical University, Shijiazhuang 050017, China
| | - YiXian Liu
- Department Of Physiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jiqing Yang
- Department Of Orthopaedic, Kingston Hospital NHS Foundation Trust, London, KT2 7QB, UK
| | - Sha Li
- Department of Human Anatomy; Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang 050017, China.
| | - Lixuan Wang
- Department Of Histology and embryology, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
37
|
Zhou J, Shou Y, Shi Q, Ye J, Li X, Zhu Z, Wang X. Fibroblast growth factor 18 attenuates renal fibrosis via AMPK/NOX4 pathway in mice. Biochem Biophys Res Commun 2025; 766:151913. [PMID: 40311293 DOI: 10.1016/j.bbrc.2025.151913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Renal fibrosis, particularly tubulointerstitial fibrosis, is a prevalent pathological process contributing to the progression of chronic kidney disease (CKD). A growing body of evidence indicates that fibroblast growth factors (FGFs) play critical roles in kidney pathophysiology. However, the role of FGF18 in the pathogenesis of kidney fibrosis and the underlying mechanisms remain unclear. In this study, we discovered a significant upregulation of FGF18 in a folic acid (FA)-induced renal fibrosis model, as well as in transforming growth factor β (TGF-β) stimulated human proximal tubular cells (HK-2 cells). Furthermore, overexpression of FGF18 in the kidney significantly alleviated FA-induced fibrosis and diminished oxidative stress. Mechanistically, FGF18 upregulated AMP-activated protein kinase (AMPK) phosphorylation via its receptor FGFR3, leading to decreased NOX4-ROS levels, reduced oxidative stress, and ultimately inhibited the expression of fibrosis-related proteins. In conclusion, our findings suggest that FGF18 attenuates FA-induced renal fibrosis, at least in partly via AMPK/NOX4 pathway.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Yanni Shou
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Qiaoyan Shi
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Junbo Ye
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Xianzhe Li
- Life Science of Pharmacy, College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, South Korea
| | - Zhongxin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China.
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
38
|
He Z, Xu Y, Zhang Y, Jin M, Sun Y, Tang F, Qiu C, Junior AMA, Cai Y, Xu X, Chen X, Chen K, Xiang G, Xiao J, Wang J, Wang J, Chen B. Betulinic acid enhances autopahgy to promote microglial M2 polarization and alleviate inflammation via AMPK-HDAC5-KLF2 signaling pathways in spinal cord injury. Int Immunopharmacol 2025; 158:114889. [PMID: 40388862 DOI: 10.1016/j.intimp.2025.114889] [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: 01/14/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Spinal cord injury (SCI) leads to neuroinflammation and activates microglia, which are crucial contributors to neurological deficits. Betulinic acid (BA), a naturally occurring pentacyclic triterpenoid, has demonstrated effectiveness in treating inflammatory and neurological disorders. This study aims to explore the potential role and underlying mechanism of BA in modulating microglial activation and inflammation in the context of SCI. Using a mouse SCI model, we assessed motor recovery via Basso Mouse Scale (BMS) and neuronal survival via H&E/Nissl staining. Western blotting, qPCR, immunofluorescence, and flow cytometry were employed to analyze microglial polarization, autophagy, and AMPK-HDAC5-KLF2 signaling in vivo and in LPS-stimulated BV2 cells. Our findings reveal that BA significantly enhances functional recovery and reduces neuronal apoptosis following SCI. Furthermore, BA facilitates the phenotypic transition of microglia from the M1 to M2 phenotype, thereby decreasing inflammatory factors in both the SCI model and LPS-stimulated BV2 cells. BA treatment restores the disrupted autophagy flux in microglia induced by SCI or LPS, which in turn mitigates M1 polarization and inflammation. Mechanistically, BA restores autophagy flux by activating the AMPK-HDAC5-KLF2 axis, thereby shifting microglia from pro-inflammatory M1 to anti-inflammatory M2 phenotype.
Collapse
Affiliation(s)
- Zili He
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yitie Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yu Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Mengqi Jin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yinuo Sun
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Fangying Tang
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chuangqi Qiu
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Abass Mashud Akinfemi Junior
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yunhao Cai
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaodan Xu
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xianghang Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Kongbin Chen
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Guangheng Xiang
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Jian Xiao
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China; Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Jian Wang
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Jing Wang
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Baoyi Chen
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
39
|
Gu J, Ruan J, Guo C, Li Z, Fu H, Xie Y, Xie H, Gong X, Shi H. Organelles Ca 2+ redistribution contributes to cadmium-induced EMT of renal cancer cells through p-cPLA 2-mediated arachidonic acid release. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118317. [PMID: 40383071 DOI: 10.1016/j.ecoenv.2025.118317] [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: 02/24/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Cadmium ion (Cd2+) is a non-essential metal that can increase cancer risk, including potentially renal cell carcinoma (RCC), though this link is not definitive. Cd2+ exposure impairs fatty acid metabolism in the kidneys, particularly affecting arachidonic acid (AA) levels, which are crucial for health. Previous studies have suggested that Cd2+-altered the AA metabolism associates with renal dysfunction. However, the role and mechanism of Cd2+-regulated AA source in promoting RCC progression are still unclear. This study aims to investigate how Cd2+ exposure affects AA levels in renal cancer cells and its role in promoting cell migration. Cd2+ exposure increases AA levels through cPLA2-mediated release. It also induces calcium ion (Ca2+) redistribution from the endoplasmic reticulum (ER) to mitochondria, activating the p38 MAPK/cPLA2 signaling pathway, and epithelial-mesenchymal transition (EMT) of Caki-1 cells. Cd2+-induced ER Ca2+ release, p38 MAPK/cPLA2 signaling activation, AA levels, and EMT of Caki-1 cells were effectively reversed by siRNA knockdown of IP3R. Both exogenous AA treatments and Cd2+-induced AA metabolite PGD2 promoted EMT and cell migration of Caki-1 cells. This study highlights Cd2+'s impact on fatty acid metabolism and organelle function in renal cancer cells, identifying potential therapeutic targets for RCC.
Collapse
Affiliation(s)
- Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China.
| | - Jiacheng Ruan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Chuanzhi Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Zehua Li
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Huilin Fu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Yimin Xie
- Affiliated Hospital of Jiangsu University-Yixing Hospital, Yixing, Jiangsu 214200, China
| | - Hebing Xie
- Jiangsu Shenhou Pharmaceutical Research Co., Ltd., Nantong, 226133, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang 212013, China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China.
| |
Collapse
|
40
|
Chen H, Liu B, Xu P, Wang H, Guo X, Liu G, Yuan J. Mechanistic role of environmental toxicants in inducing cellular ferroptosis and its associated diseases. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118269. [PMID: 40344778 DOI: 10.1016/j.ecoenv.2025.118269] [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: 08/26/2024] [Revised: 03/04/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Due to exposure factors such as industrial exhaust, sewage discharge, pesticide runoff, automobile exhaust, and fuel combustion, environmental toxicants are widely present in daily life. Organisms are exposed to these environmental toxicants through contaminated air, food, and drinking water, and these environmental toxicants enter the human body and cause cytotoxicity and diseases through various pathways. As a new cell death mode that is different from cell necrosis, apoptosis, and autophagy, ferroptosis are mainly dysregulation of intracellular iron metabolism, lipid metabolism disorders, and the dysregulation of the antioxidant defense system, leading to lipid peroxidation and ultimately to the rupture of the cell membrane, damage, and cell death. Studies have shown that environmental toxicants induce a series of diseases, such as digestive diseases, urinary diseases, respiratory diseases, neurological disorders, and reproductive diseases, through the above mechanisms. We elaborate the mechanism of common environmental toxicants in inducing ferroptosis and the related systemic diseases mediated through the ferroptosis to provide the theoretical basis for preventing and treating environmental toxicant-related diseases. Nonetheless, our understanding of ferroptosis remains incomplete. For example, mechanisms and methods for the selective control of ferroptosis remain elusive, elucidating these mechanisms and strategies may be critical for leveraging knowledge of ferroptosis to treat related diseases.
Collapse
Affiliation(s)
- Hong Chen
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Bingchun Liu
- Stem Cell Laboratory; Central Laboratory of Organ Transplantation;Inner Mongolia Autonomous Region Engineering Laboratory for Genetic Test and Research of Tumor Cells, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Peixin Xu
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Huizeng Wang
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Xin Guo
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Gang Liu
- Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Jianlong Yuan
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China.
| |
Collapse
|
41
|
Liu R, Xi Y, Duan X, Zhao Y, Tian Z. Exerkine-mediated organ interactions: A new interpretation of exercise on cardiovascular function improvement. Life Sci 2025; 371:123628. [PMID: 40210118 DOI: 10.1016/j.lfs.2025.123628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Cardiovascular diseases impair the structure and function of distal organs, including the liver, skeletal muscle, kidney, and adipose tissue. Exercise stimulates the interaction between the cardiovascular system and distal organs that is important for disease rehabilitation and organ health. However, the mechanisms by which exercise improves cardiovascular function through exerkine-mediated organ crosstalk remain incompletely elucidated. We used cardiovascular, exercise, exerkines, skeletal muscle, liver, kidney, and adipose tissue as keywords to search for the relevant articles, sorted out the differences between different exercise types, summarized the functions of 17 exerkines, focused on reviewing and categorizing the molecular mechanisms of interactions between the cardiovascular system and remote organs. We also look forward to future research perspectives on exercise prevention and control of chronic metabolic diseases. The aim of this review is to provide a new theoretical basis for establishing clinical rehabilitation and exercise prescriptions for cardiovascular system diseases.
Collapse
Affiliation(s)
- Renhan Liu
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China
| | - Yue Xi
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China..
| | - Xinyan Duan
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China
| | - Yifei Zhao
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China
| | - Zhenjun Tian
- Laboratory of Exercise Intervention on Metabolic Syndrome, Brain-Heart Health and Education, Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi' an 710119, PR China..
| |
Collapse
|
42
|
Li GZ, Liu JY, Zhou H. Ferroptosis: A novel therapeutic target for diabetic cardiomyopathy. World J Diabetes 2025; 16:104665. [DOI: 10.4239/wjd.v16.i6.104665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/24/2025] [Accepted: 05/07/2025] [Indexed: 06/13/2025] Open
Abstract
Ferroptosis is a new type of programmed cell death caused by the accumulation of iron-dependent lipid peroxides, and it plays a role in the occurrence and progression of diverse diseases. Diabetic cardiomyopathy (DCM), a serious cardiovascular complication in patients with diabetes, eventually progresses to refractory heart failure (HF), which increases the risk of hospitalization for HF and cardiovascular death in patients with diabetes. Despite glycemic control, effective strategies to prevent DCM onset are currently lacking. Accumulating evidence suggests that ferroptosis is involved in oxidative stress, inflammation, and abnormal autophagy in diabetic myocardium, which plays an important role in myocardial apoptosis, hypertrophy, and cardiac fibrosis. The inhibition of ferroptosis can relieve DCM. Presently, ferroptosis inhibitors have been broadly suggested for the treatment of iron overload-related cardiomyopathy. This article reviewed relevant studies to offer a new therapeutic target for DCM.
Collapse
Affiliation(s)
- Gui-Zhi Li
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Jia-Yin Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Hong Zhou
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
43
|
Chen JH, Dai XC, Quan ZJ, Liu XY. Erianin mitigates diabetic cardiomyopathy via adenosine monophosphate-activated protein kinase-nuclear factor erythroid 2-related factor 2-heme oxygenase-1 pathway activation. World J Diabetes 2025; 16:103685. [DOI: 10.4239/wjd.v16.i6.103685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/21/2025] [Accepted: 04/17/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Erianin is a natural bibenzyl compound extracted from Dendrobium chrysotoxum and is known for its anti-inflammatory and antioxidant properties.
AIM To explore the possible therapeutic mechanisms of erianin and determine if it can reduce cardiac damage in mice with type 2 diabetes.
METHODS High-fat diet and intraperitoneal injections of streptozotocin were used to induce type 2 diabetes mellitus in C57BL/6 mice. Mice were divided into different groups including control, model, and treatment with various doses of erianin (10, 20, and 40 mg/kg) as well as ML-385 + erianin group.
RESULTS Erianin reduced oxidative stress and inflammation and alleviated diabetic cardiomyopathy through the activation of the adenosine monophosphate-activated protein kinase (AMPK)-nuclear factor erythroid 2-related factor 2 (Nrf2)-heme oxygenase-1 (HO-1) pathway. Treatments with erianin-M and erianin-H promoted weight stabilization and normalized fasting glucose levels relative to diabetic controls. Echocardiographic assessment demonstrated that erianin dose-dependently enhanced left ventricular systolic function (left ventricular ejection fraction, left ventricular fractional shortening) and mitigated ventricular remodeling (left ventricular internal diameter at end-diastole, left ventricular internal diameter at end-systole; P < 0.05 vs model group). No significant differences were observed between the ML-385 + erianin and placebo-treated groups. Histopathological examination through hematoxylin-eosin staining indicated that erianin ameliorated myocardial fiber fragmentation, structural disorganization, inflammatory cell infiltration, and cytolytic damage. Furthermore, it significantly reduced the serum levels of cardiac troponin I, creatine kinase, and its MB isoenzyme. However, the ML-385 + erianin co-treatment failed to alleviate myocardial injury. Metabolic profiling revealed erianin-mediated improvements in glycemic regulation (glycated hemoglobin: P < 0.001), plasma insulin homeostasis, and lipid metabolism (total cholesterol, triglycerides, low-density lipoprotein cholesterol reduction, and high-density lipoprotein cholesterol restoration; P < 0.05 vs model group). Proinflammatory cytokines including tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 were markedly suppressed in the erianin-M and erianin-H groups compared with the model group, whereas no significant differences were detected between the model and ML-385 + erianin groups. Oxidative stress parameters showed decreased malondialdehyde levels accompanied by elevated superoxide dismutase and catalase activities in erianin-treated groups, with the most pronounced effects in the erianin-H group (P < 0.05). Western blot analysis confirmed the significant upregulation of proteins associated with the AMPK/Nrf2/HO-1 pathway in erianin-M and erianin-H groups. These protective effects were abolished in the ML-385 + erianin co-treatment group, which showed no statistical differences from the model group.
CONCLUSION Erianin can effectively alleviate myocardial injury in type 2 diabetic mice by activating the AMPK-Nrf2-HO-1 pathway.
Collapse
Affiliation(s)
- Jia-Hui Chen
- Department of Gerontology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121012, Liaoning Province, China
- Department of Gerontology, The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorder, Wenzhou 325000, Zhejiang Province, China
| | - Xiao-Chun Dai
- Department of Gerontology, The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental Disorder, Wenzhou 325000, Zhejiang Province, China
| | - Zi-Jiao Quan
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, Zhejiang Province, China
| | - Xin-Yu Liu
- Department of Gerontology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121012, Liaoning Province, China
| |
Collapse
|
44
|
Xing QC, Chen J, Liu Z, Li WC, Liu X, Li W. Autophagy in Schwann cells: A potential pharmacotherapeutic target in diabetic peripheral neuropathy. World J Diabetes 2025; 16:105709. [DOI: 10.4239/wjd.v16.i6.105709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/27/2025] [Accepted: 05/16/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes and is characterized by sensory and motor impairments resulting from neural injury. Schwann cells (SCs), which are important for peripheral nerve function, are compromised under hyperglycemic conditions, leading to impaired axonal regeneration and demyelination. Autophagy, a cellular degradation process, is essential for SC function and significantly influences DPN progression. This article highlights the significance of autophagy in SCs and its potential as a pharmacotherapeutic target in DPN. We discuss the mechanisms of autophagy in SCs, including the mammalian target of rapamycin, adenosine monophosphate-activated protein kinase, and phosphatase and tensin homolog-induced putative kinase/parkin pathways, and their dysregulation in DPN. This article also examines various natural products and chemical agents that modulate autophagy and enhance the efficacy of DPN treatment. These agents target key signaling pathways, such as adenosine monophosphate-activated protein kinase/mammalian target of rapamycin and demonstrate potential in promoting nerve regeneration and restoring SC function. The roles of exosomes, long non-coding RNA, and proteins in the regulation of autophagy have also been explored. In conclusion, targeting autophagy in SCs is a promising strategy for DPN treatment and offers new insights into therapeutic interventions. Further research is warranted to fully exploit these targets for clinical applications.
Collapse
Affiliation(s)
- Qi-Chang Xing
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Jia Chen
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Wen-Can Li
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Wei Li
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| |
Collapse
|
45
|
Li YW, Tu SX, Li ZX, Ding YQ, Hu L. Manifold functions of Mediator complex in neurodevelopmental disorders. Neurobiol Dis 2025; 210:106913. [PMID: 40246246 DOI: 10.1016/j.nbd.2025.106913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025] Open
Abstract
Neurodevelopmental disorders (NDDs) encompass a diverse range of impairments affecting brain development and functions, often presenting as deficits in motor skills, cognitive abilities, language development and neuropsychiatric health. The emergence of next-generation sequencing has unveiled numerous genetic variants linked to NDDs, implicating molecular pathways involved in essential neuronal processes such as synaptic plasticity, neuronal architecture and proteostasis. Central to these processes is the Mediator complex, a highly conserved multi-subunit assembly crucial for RNA polymerase II (Pol II)-dependent transcription. The Mediator functions as a key regulator of gene expression, playing a pivotal role in coordinating cellular processes essential for neuronal differentiation and developmental signaling cascades. Increasingly evidence has shown that its dysfunction is highly associated with the pathogenesis of NDDs. This review aims to comprehensively examine the structural and functional characteristics of individual mediator subunits. We will focus on clinical case reports and recent preclinical studies that highlight the connection between genetic abnormalities in the Mediator complex and specific neurodevelopmental phenotypes, ultimately guiding the development of enhanced diagnostic tools and therapeutic interventions. Furthermore, this review will advance our understanding of the general role transcriptional regulation plays in the etiology of NDDs.
Collapse
Affiliation(s)
- Yi-Wei Li
- Laboratory Animal Center, Fudan University, Shanghai 200032, China
| | - Si-Xin Tu
- Laboratory Animal Center, Fudan University, Shanghai 200032, China
| | - Ze-Xuan Li
- Laboratory Animal Center, Fudan University, Shanghai 200032, China
| | - Yu-Qiang Ding
- Laboratory Animal Center, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, Fudan University, Shanghai 200032, China.
| | - Ling Hu
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Laboratory Animal Center, Fudan University, Shanghai 200032, China.
| |
Collapse
|
46
|
Zhu M, Liu W, Su S, Gong M, Liao G, Fu F, Chen G, Rao Z, Cheng J, Liu J, Lu Y, Chen Y. Reprogramming of lipids and amino acids metabolism is an early event in myocardium of type 1 diabetic rhesus monkeys. J Pharm Biomed Anal 2025; 258:116699. [PMID: 39914331 DOI: 10.1016/j.jpba.2025.116699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/10/2025]
Abstract
Diabetic cardiomyopathy (DC) refers to the abnormal myocardial structure and performance induced by diabetes. Although numerous studies have been carried out, the pathophysiological mechanisms of cardiovascular disorders during diabetes have not been fully clarified. Here, we compared the cardiomyopathy of healthy rhesus monkeys and rhesus monkeys with a history of streptozocin induced type 1 diabetes (T1D) over 7 years. Through comparing the cardiac function using echocardiography, and detecting the serum biochemical indexes, and changes of left ventricle (LV), we found that decreased systolic function, higher blood glycosylated hemoglobin A1c (HbA1C) level, hyperglycemia, and hyperlipidemia were early events in diabetic rhesus monkeys. In addition, cardiac histological analysis showed mildly fibrosis and early myocardial hypertrophy, as evidenced by increased Sirius red stained area and cross-sectional area of left ventricle. Transcriptome results revealed that the nutrients metabolism and extracellular matrix related pathways were markedly changed in the left ventricle of diabetic monkeys. Targeted metabolomics and targeted lipid metabolomics further revealed that disturbed amino acid metabolism and lipid accumulation in the LV of diabetic monkeys manifested by accumulated branched chain amino acids (BCAAs) and triglycerides (TAGs), and reduced contents of sphingolipids, glycerophospholipids, cholesteryl esters and carnitines. In conclusion, we reported here for the first time that diabetes lasting for more than 7 years leads to some early pathological changes of myocardium in rhesus monkeys. The cardiac function is mildly compromised and the reprogramming of lipids and amino acids metabolism might play important roles in the progression of DC.
Collapse
Affiliation(s)
- Min Zhu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wen Liu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Shan Su
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Meng Gong
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, PR China; Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Guangneng Liao
- Animal experimental center of West China hospital, Sichuan University, Chengdu, PR China
| | - Fudong Fu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, PR China
| | - Gen Chen
- Development and Application of Human Major Disease Monkey Model Key Laboratory of Sichuan Province, Sichuan Hengshu Bio-Technology Co. Ltd., Sichuan 644600, PR China
| | - Zhiyong Rao
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jingqiu Cheng
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jingping Liu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yanrong Lu
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Younan Chen
- Department of Clinical Nutrition and NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China; Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610000, PR China.
| |
Collapse
|
47
|
Wang YB, Li ZP, Wang P, Wang RB, Ruan YH, Shi Z, Li HY, Sun JK, Mi Y, Li CJ, Zheng PY, Zhang CJ. Iron dysregulation, ferroptosis, and oxidative stress in diabetic osteoporosis: Mechanisms, bone metabolism disruption, and therapeutic strategies. World J Diabetes 2025; 16:106720. [DOI: 10.4239/wjd.v16.i6.106720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 04/16/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic osteoporosis (DOP) is a common complication in diabetes, driven by hyperglycemia-induced metabolic disturbances, chronic inflammation, and oxidative stress. This review describes the critical role of iron metabolism dysregulation in DOP pathogenesis, focusing on ferroptosis, a novel iron-dependent cell death pathway characterized by lipid peroxidation and reactive oxygen species (ROS) overproduction. Diabetic conditions exacerbate iron overload, impairing osteoblast function and enhancing osteoclast activity, while triggering ferroptosis in bone cells. Ferroptosis not only accelerates osteoblast apoptosis but also amplifies osteoclast-mediated bone resorption, synergistically promoting bone loss. Furthermore, chronic inflammation and oxidative stress disrupt the balance between bone formation and resorption, with elevated pro-inflammatory cytokines (e.g., tumor necrosis factor-α, interleukin-6) and ROS exacerbating cellular dysfunction. Therapeutic strategies targeting iron metabolism (e.g., deferoxamine) and ferroptosis inhibition (e.g., nuclear factor erythroid 2-related factor 2/heme oxygenase-1 pathway activation, antioxidants like melatonin) demonstrate potential to mitigate DOP progression. Future research should prioritize personalized interventions, clinical trials of iron chelators and antioxidants, and mechanistic studies to refine therapeutic approaches. This review provides a comprehensive framework for understanding DOP pathogenesis and highlights innovative strategies to improve bone health in diabetic patients.
Collapse
Affiliation(s)
- Yao-Bin Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhi-Peng Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Peng Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Rui-Bo Wang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yu-Hua Ruan
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhen Shi
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hao-Yu Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jin-Ke Sun
- The Third Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Cheng-Jin Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Peng-Yuan Zheng
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Chang-Jiang Zhang
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
48
|
Domingues C, Jarak I, Matos A, Veiga F, Vitorino C, Dourado M, Figueiras A. Unraveling rosmarinic acid anticancer mechanisms in oral cancer malignant transformation. Eur J Pharmacol 2025; 997:177466. [PMID: 40064225 DOI: 10.1016/j.ejphar.2025.177466] [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: 01/17/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Oral squamous cell carcinoma (OSCC) is expected to rise ca. 40 % by 2040. Rosmarinic acid (RA) has been recognized for its anticancer properties, although its role in OSCC has been neglected. This work exploits the activity of RA in 2D and 3D models of OSCC cells to compel a roadmap for its anticancer properties. The results demonstrated that RA significantly reduced cell mass and metabolic activity in a dose, time, and cell-type-dependent manner, predominantly in highly-invasive OSCC, without compromising normal mucosa in therapeutic doses. RA decreased mitochondria membrane potential and increased redox state, which was corroborated by pioneering observations on the metabolome landscape of OSCC cells (glutathione reduction and acetate and fumarate release). RA triggered autophagy, upregulating BNIP3 and BCNL1 and downregulating BIRC5. The upregulation of CADM1 and downregulation of VIM, CADM2, SNAIL1, and SOX9 highlighted the modulation of epithelial-mesenchymal transition and the remodeling of the extracellular matrix by the downregulation of MMP-2 and MMP-9. RA interacts with P-glycoprotein with the highest docking score of -6.4 kcal/mol. The HSC-3 cell surface charge decreased after RA treatment (-22.6 ± 0.3 mV vs. -26.3 ± 0.3 mV, p < 0.0001), suggesting a reversion of cell polarity and the impairment of invasion. RA also shrank the growth and the metabolic activity of multicellular tumor spheroids. Its modest protein binding with human saliva sheds light on its administration by the oromucosal route. Overall, this work supports the need for further research on the anticancer potential of RA in OSCC, either in monotherapy, combined with conventional treatments, or conveyed in nanosystems.
Collapse
Affiliation(s)
- Cátia Domingues
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; REQUIMTE/LAQV, Drug Development and Technologies Laboratory, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ivana Jarak
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Ana Matos
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; Chemical Engineering and Renewable Resources for Sustainability, CERES, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Francisco Veiga
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; REQUIMTE/LAQV, Drug Development and Technologies Laboratory, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Carla Vitorino
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Marília Dourado
- Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; REQUIMTE/LAQV, Drug Development and Technologies Laboratory, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
49
|
Cannarella R, Rubulotta M, Cannarella V, La Vignera S, Calogero AE. A holistic view of SGLT2 inhibitors: From cardio-renal management to cognitive and andrological aspects. Eur J Intern Med 2025:S0953-6205(25)00247-X. [PMID: 40517121 DOI: 10.1016/j.ejim.2025.06.010] [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: 02/19/2025] [Revised: 06/04/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial disease associated with complications that significantly affect both survival and quality of life, including cardiovascular, renal, cognitive, sexual, and reproductive dysfunctions. Sodium-glucose cotransporter 2 (SGLT2) inhibitors (SGLT2is) have emerged as a transformative class of drugs, demonstrating benefits that extend beyond glycemic control. Large clinical trials have shown that SGLT2is reduce hospitalization for heart failure by 25-35% and slow progression of chronic kidney disease by 30-45%, with variation based on the specific agent, dose, and patient population. This narrative review examines not only these well-established benefits but also emerging evidence regarding their effects in less-explored domains. SGLT2is have been associated with improved cognitive performance, potentially through reductions in neuroinflammation and oxidative stress. In the sexual and reproductive domains, studies in men with diabetes mellitus suggest potential benefits of SGLT2is in improving erectile function, sperm motility, and testosterone levels, likely mediated by antioxidant and anti-inflammatory mechanisms. By integrating current evidence across multiple systems, this review emphasizes the role of SGLT2is in a holistic, multidisciplinary approach to the management of patients with T2DM.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Michele Rubulotta
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vittorio Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Dipartimento di Scienze Mediche, IOC di Medicina Generale, AOU Policlinico "G. Rodolico-San Marco", University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
50
|
Casetti R, Ciccosanti F, Lamsira HK, Pinnetti C, Mazzotta V, Ciolfi S, Sacchi A, Amendola A, Ippolito G, Piacentini M, Nardacci R. Autophagy is influenced by vitamin D 3 level in people with HIV-1. Biol Direct 2025; 20:69. [PMID: 40514685 DOI: 10.1186/s13062-025-00660-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Autophagy is the primary catabolic process responsible for degrading intracellular components and potentially harmful cytosolic entities by delivering them to lysosomes. Notably, this mechanism is crucial for controlling intracellular pathogens, with significant implications for both innate and adaptive immunity. In the context of HIV-1 infection, emerging evidence suggests that autophagy contributes to immune responses against the virus. Various compounds can modulate autophagy, among which vitamin D₃ is particularly effective due to its ability to prevent inflammation and slow HIV-1 disease progression. Indeed, vitamin D₃ contributes to regulating both innate and adaptive immunity, thereby modulating antiviral and antibacterial inflammatory responses. Importantly, vitamin D₃ deficiency is highly prevalent among people with HIV (PWH) and has been associated with an increased risk of severe disease progression. RESULTS In this study, we investigated the relationship between serum vitamin D₃ levels and the expression of autophagy markers in peripheral blood mononuclear cells from different categories of PWH: PWH under antiretroviral therapy (ART) with either normal vitamin D₃ levels or hypovitaminosis, and treatment-naïve PWH with either normal vitamin D₃ levels or hypovitaminosis. Our results show that low vitamin D₃ blood levels is associated with lower expression of the main factors involved in the autophagy mechanism, particularly in treatment-naïve PWH. CONCLUSIONS Our findings suggest that normal blood level of vitamin D₃ may play a crucial role in promoting autophagy in PWH. The observed differences in autophagy-related protein expression between ART-treated and untreated individuals underscore the intricate relationship between vitamin D₃ levels, ART exposure, and autophagic regulation. This is a preliminary exploration of the effects of vitamin D₃ on autophagy in PWH. Further studies are needed to deepen and explore the interplay between vitamin D₃ and autophagy in greater depth. A better understanding of these mechanisms could help to develop novel therapeutic strategies aimed at mitigating immune depletion and chronic inflammation, ultimately improving clinical outcomes for individuals living with HIV.
Collapse
Affiliation(s)
- Rita Casetti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - IRCCS, Rome, 00149, Italy
| | - Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - IRCCS, Rome, 00149, Italy
| | - Harpreet Kaur Lamsira
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Rome, 00131, Italy
| | - Carmela Pinnetti
- Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, 00149, Italy
| | - Valentina Mazzotta
- Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, 00149, Italy
| | - Serena Ciolfi
- Department of Science, University of Roma Tre, Rome, 00146, Italy
| | | | - Alessandra Amendola
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases 'Lazzaro Spallanzani'- IRCCS, Rome, 00149, Italy
| | - Giuseppe Ippolito
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Rome, 00131, Italy
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - IRCCS, Rome, 00149, Italy
- Department of Biology, University of Rome 'Tor Vergata', Rome, 00133, Italy
| | - Roberta Nardacci
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Rome, 00131, Italy.
| |
Collapse
|