1
|
Choi JC. Perinuclear organelle trauma at the nexus of cardiomyopathy pathogenesis arising from loss of function LMNA mutation. Nucleus 2025; 16:2449500. [PMID: 39789731 PMCID: PMC11730615 DOI: 10.1080/19491034.2024.2449500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Over the past 25 years, nuclear envelope (NE) perturbations have been reported in various experimental models with mutations in the LMNA gene. Although the hypothesis that NE perturbations from LMNA mutations are a fundamental feature of striated muscle damage has garnered wide acceptance, the molecular sequalae provoked by the NE damage and how they underlie disease pathogenesis such as cardiomyopathy (LMNA cardiomyopathy) remain poorly understood. We recently shed light on one such consequence, by employing a cardiomyocyte-specific Lmna deletion in vivo in the adult heart. We observed extensive NE perturbations prior to cardiac function deterioration with collateral damage in the perinuclear space. The Golgi is particularly affected, leading to cytoprotective stress responses that are likely disrupted by the progressive deterioration of the Golgi itself. In this review, we discuss the etiology of LMNA cardiomyopathy with perinuclear 'organelle trauma' as the nexus between NE damage and disease pathogenesis.
Collapse
Affiliation(s)
- Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Zhu M, Xiang W, Zhu Z, Nie B, Zhen X, Chen C, Wang T. Bibliometric analysis of autophagy in the diagnosis and treatment of osteosarcoma: a bibliometric analysis (2007-2023). Cancer Biol Ther 2025; 26:2484825. [PMID: 40146196 DOI: 10.1080/15384047.2025.2484825] [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: 04/10/2024] [Revised: 12/14/2024] [Accepted: 03/23/2025] [Indexed: 03/28/2025] Open
Abstract
Osteosarcoma is the most common primary bone tumor in children and adolescents. Its pathogenesis is complex and poses difficulties in treatment. Autophagy is a cell biological process that plays a crucial role in the mechanistic study and treatment of osteosarcoma. The objective of this study is to evaluate the past research progress from 2007 to 2023 and visualize the key research directions through bibliometric methods. Relevant publications published from the start of 2007 to the end of 2023 were searched and screened in the Web of Science Core Collection. They were analyzed and visualized using CiteSpace and the Bibliometric online analysis platform in terms of country, institution, author, journal, cited references, and keywords. In total, 619 publications from 522 journals with 682 authors from 42 countries were screened. The country with the highest number of publications is China (n = 445, 71.890%), followed by the United States (n = 60, 9.693%). The research institution with the highest number of publications is Shanghai Jiao Tong University (n = 42, 6.785%). The author with the highest number of publications is Cai, Zhengdong (n = 7), while the most cited author is Mizushimma N (n = 93). Among many journals, AUTOPHAGY has the most citations (n = 342), while CANCER LETT shows the greatest centrality (Centrality = 0.05). "Autophagy" is the most cited keyword (n = 177), and the keyword with the largest burst intensity is "cancer cells" (Strength = 6.27), which lasted from 2011 to 2014. China is a major contributor to autophagy research in the field of osteosarcoma, followed by the United States. All publications are in high-quality journals. "Autophagy" is a hot research topic in this field.
Collapse
Affiliation(s)
- Min Zhu
- Department of Anesthesiology, The Third Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wei Xiang
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhoujun Zhu
- Department of Joint Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bingjie Nie
- Department of Anesthesiology, The Third Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xinyue Zhen
- Department of Anesthesiology, The Third Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Chen Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tianhai Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
3
|
Ye K, Zhao X, Liu L, Ge F, Zheng F, Liu Z, Tian M, Han X, Gao X, Xia Q, Wang D. Comparative Analysis of Human Brain RNA-seq Reveals the Combined Effects of Ferroptosis and Autophagy on Alzheimer's Disease in Multiple Brain Regions. Mol Neurobiol 2025; 62:6128-6149. [PMID: 39710824 DOI: 10.1007/s12035-024-04642-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024]
Abstract
Ferroptosis and autophagy are closely associated with Alzheimer's disease (AD). Elevated ferric ion levels can induce oxidative stress and chronic inflammatory responses, resulting in brain tissue damage and further neurological cell damage. Autophagy in Alzheimer's has a dual role. On one hand, it protects neurons by removing β-amyloid and cellular damage products caused by oxidative stress and inflammation. On the other hand, abnormal autophagy is linked to neuronal apoptosis and neurodegeneration. However, the intricate interplay between ferroptosis and autophagy in AD remains insufficiently explored. This study focuses on the roles of ferroptosis and autophagy in AD and their interconnection through bioinformatics analysis, shedding light on the disease. Ferroptosis and autophagy significantly correlate with the development and course of AD. Using PPI network analysis and unsupervised consistency clustering analysis, we uncovered a complex network of interactions between ferroptosis and autophagy during disease progression, demonstrating a significant congruence in their modification patterns. Functional analyses further demonstrated that ferroptosis and autophagy together affect the immunological status and synaptic regulation in hippocampal regions in patients with AD, which significantly impacts the start and progression of the disease.
Collapse
Affiliation(s)
- Ke Ye
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Zijie Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Mengjie Tian
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Xinyu Han
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China.
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, 150000, Heilongjiang, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150000, Heilongjiang, China.
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China.
| |
Collapse
|
4
|
Chen Y, Shu C, Yan Z, Zhang S, Zhang W, Zhao J, Wang A, Li J, Zeng Y, Zhu J, Huang JA, Liu Z. Liensinine overcomes EGFR-TKI resistance in lung adenocarcinoma through DRP1-mediated autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156593. [PMID: 40054177 DOI: 10.1016/j.phymed.2025.156593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Persistent upregulation of autophagy contributes to tumour cells' resistance to EGFR-TKI therapy, and hence, inhibiting autophagy could be a valuable strategy for overcoming such resistance. OBJECTIVES This study investigated the effects of liensinine in EGFR-TKI resistant lung adenocarcinoma (LUAD) and to explore the underlying mechanism. METHODS CCK-8 assay, colony formation, EdU assay and apoptosis assays were conducted for investigating the effect of EGFR-TKI and liensinine combination treatment in LUAD. Furthermore, autophagic flux were detected by western blot, fluorescence assays and TEM. In addition, by employing a DARTS approach, a CETSA assay, and SPR analysis, we identified DRP1 as a target of liensinine. Finally, by establishing a xenograft model of the disease, the impact of combination treatment in vivo was assessed. RESULT In vitro and in vivo experiments revealed that the novel autophagy inhibitor liensinine enhanced the sensitivity of LUAD to EGFR-TKIs. This effect was achieved by inhibiting autophagic flux. We then examined whether liensinine inhibits autophagic flux through the impairment of autophagosome and autolysosome degradation. Furthermore, we identified DRP1 as a target of liensinine. The activation of DRP1 by liensinine through dephosphorylation at Ser637 promotes the accumulation of autophagosomes and autolysosomes while simultaneously blocking autophagic flux, thereby enhancing the cancer cell-killing effects of EGFR-TKIs. CONCLUSIONS Our study validated the efficacy of liensinine in overcoming EGFR-TKI resistance and elucidated the mechanism underlying liensinine's inhibition of autophagy.
Collapse
Affiliation(s)
- Yuling Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Chenying Shu
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhaowei Yan
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Saiqun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Weijie Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jian Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Anqi Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jianjun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Jian-An Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China; Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.
| |
Collapse
|
5
|
Suman TY, Kwak IS. Current understanding of human bioaccumulation patterns and health effects of exposure to perfluorooctane sulfonate (PFOS). JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137249. [PMID: 39842114 DOI: 10.1016/j.jhazmat.2025.137249] [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: 10/16/2024] [Revised: 12/24/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant of global concern due to its environmental presence,bioaccumulative potential and toxicological impacts. This review synthesizes current knowledge regarding PFOS exposure, bioaccumulation patterns and adverse health outcomes in human population. Analysis of worldwide biomonitoring data, and epidemiological studies reveals PFOS systemic effects, including immunological dysfunction (decreased vaccine response), developmental toxicity (reduced birth weight), hepatic metabolic disruption, potential carcinogenogenicity, and reproductive abnormalities. At the molecular level, PFOS induces toxicity through multiple pathways, including PI3K/AKT/mTOR pathway inhibition, PPARα activation, NF-κB signaling modulation, and oxidative stress induction. Recent advances in analytical methodologies have enhanced our understanding of PFOS distribution and fate, while evolving egulatory frameworks attempts to address its risk. This review identifies critical research gaps and emphasized the need for coordinated multidisciplinary approaches to address this persistent environmental contaminant.
Collapse
Affiliation(s)
| | - Ihn-Sil Kwak
- Fisheries Science Institute, Chonnam National University, Yeosu 59626, South Korea; Department of Ocean Integrated Science, Chonnam National University, Yeosu 59626, South Korea.
| |
Collapse
|
6
|
Guo L, Wang N, Chen J, Zhang R, Li D, Yang L. Cellular senescence and glaucoma. Exp Gerontol 2025; 202:112718. [PMID: 39983803 DOI: 10.1016/j.exger.2025.112718] [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: 11/13/2024] [Revised: 01/06/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Cellular senescence, a characteristic feature of the aging process, is induced by diverse stressors. In recent years, glaucoma has emerged as a blinding ocular disease intricately linked to cellular senescence. The principal pathways implicated are oxidative stress, mitochondrial dysfunction, DNA damage, autophagy impairment, and the secretion of various senescence- associated secretory phenotype factors. Research on glaucoma-associated cellular senescence predominantly centers around the increased resistance of the aqueous humor outflow pathway, which is attributed to the senescence of the trabecular meshwork and Schlemm's canal. Additionally, it focuses on the mechanisms underlying retinal ganglion cell senescence in glaucoma and the corresponding intervention measures. Given that cell senescence represents an irreversible phase preceding cell death, an in-depth investigation into its mechanisms in the pathogenesis and progression of glaucoma, particularly by specifically blocking the signal transduction of cell senescence, holds the potential to decrease the outflow resistance of aqueous humor. This, in turn, could provide a novel avenue for safeguarding the optic nerve in glaucoma.
Collapse
Affiliation(s)
- Liang Guo
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Na Wang
- The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Chen
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Rui Zhang
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dan Li
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lu Yang
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
7
|
Wen J, Ou SJ, Liu JB, Zeng W, Yang R, Qu YD, Li JX, Xia CL, Yang Y, Zhang W, Qi Y, Xu CP. Single-cell RNA sequencing reveals the role of immune-related autophagy in aseptic loosening of biomaterials bone-implant. BIOMATERIALS ADVANCES 2025; 169:214190. [PMID: 39842168 DOI: 10.1016/j.bioadv.2025.214190] [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: 09/14/2024] [Revised: 12/29/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
This study employed single-cell RNA sequencing (scRNA-seq) to investigate the role of immune-related autophagy in the mechanism of aseptic loosening (AL) of biomaterial bone-implant. Through single-cell analysis of AL tissues, we mapped the cellular landscape, revealing various cell types and their characteristics within the context of AL. Our study specifically targeted immune cell subpopulations, including macrophages and neutrophils. The results suggest the autophagy-related gene Ctsb was downregulated in AL, especially in macrophages. Subsequently our experiments confirmed the correlation between reduced Ctsb expression and enhanced autophagy, which may affect macrophage apoptosis and osteoblast differentiation, ultimately contributing to periprosthetic osteolysis and AL. This study offers novel perspectives into the role of immune related autophagy in the mechanism of AL and establishes a foundation for the future development of targeted therapeutic strategies for AL.
Collapse
Affiliation(s)
- JianPing Wen
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Shuan-Ji Ou
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Jia-Bao Liu
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Wei Zeng
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - RongShen Yang
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Yu-Dun Qu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Jia-Xuan Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Chang-Liang Xia
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Yang Yang
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Wei Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Yong Qi
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
| | - Chang-Peng Xu
- Department of Orthopedics, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| |
Collapse
|
8
|
Yang B, Shen M, Lu C, Wang Y, Zhao X, Zhang Q, Qin X, Pei J, Wang H, Wang J. RNF144A inhibits autophagy by targeting BECN1 for degradation during L. monocytogenes infection. Autophagy 2025; 21:789-806. [PMID: 39608349 PMCID: PMC11925115 DOI: 10.1080/15548627.2024.2429380] [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: 04/19/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Listeria monocytogenes (L. monocytogenes, Lm) is widely used in the laboratory as an infection model for the research on pathogenesis and host defense against gram-positive intracellular bacteria. Macroautophagy (called simply "autophagy" hereafter), is important in the host defense against pathogens, such as bacteria, viruses, and parasites. BECN1 plays a pivotal role in the initiation of autophagy and accumulating evidence indicates that post-translational modifications of BECN1 provide multiple strategies for autophagy regulation. In this study, we demonstrated that the RING1-IBR-RING2 (RBR) family member RNF144A (ring finger protein 144A), which was induced by Lm infection, promoted Lm infection in an autophagy-dependent but STING1-independent pattern. rnf144a deficiency in mice protected mice from Lm infection with inhibited innate immune responses. Interestingly, RNF144A decreased Lm-induced autophagosome accumulation. Mechanistic investigation indicated that RNF144A interacted with BECN1 and promoted its K48-linked ubiquitination, leading to the subsequent proteasome-dependent degradation of BECN1 and reduced autophagosome accumulation. Further study demonstrated that RNF144A promoted the ubiquitination of BECN1 at K117 and K427, and these two ubiquitination sites were essential to the role of BECN1 in autophagy and Lm infection. Thus, our findings suggested a new regulator in intracellular bacterial infection and autophagy, which may contribute to our understanding of host defense against intracellular bacterial infection via autophagy.Abbreviations: ATG3: autophagy related 3; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG10: autophagy related 10; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; Baf A1: bafilomycin A1; BECN1: beclin 1; BMDC: bone marrow-derived dendritic cell; BMDM: bone marrow-derived macrophage; CFUs: colony-forming units; CHX: cycloheximide; CQ: chloroquine; CXCL10/IP-10: C-X-C motif chemokine ligand 10; EBSS: Earle's balanced salt solution; ELISA: enzyme-linked immunosorbent assay; IFIT1/ISG56: interferon induced protein with tetratricopeptide repeats 1; IFNB/IFN-β: interferon beta; IL6: interleukin 6; IRF3, interferon regulatory factor 3; Lm: L. monocytogenes; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MOI: multiplicity of infection; PLA: proximity ligation assay; PMA: phorbol myristate acetate; PMA-THP1, PMA-differentiated THP1; PMs: peritoneal macrophages; PTMs: posttranslational modifications; RBR: RING1-IBR-RING2; RNF144A: ring finger protein 144A; STING1, stimulator of interferon response cGAMP interactor 1; TBK1, TANK binding kinase 1; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Bo Yang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Mengyang Shen
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chen Lu
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yi Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xin Zhao
- Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qunmei Zhang
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Xiao Qin
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinyong Pei
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jie Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
9
|
Li B, Yang XM, Zhou XM, Huang YQ. Effect of pyrroloquinoline quinone on skin aging in Bmi-1 KO mice and underlying mechanisms. PLoS One 2025; 20:e0319770. [PMID: 40153371 DOI: 10.1371/journal.pone.0319770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/09/2025] [Indexed: 03/30/2025] Open
Abstract
To investigate the effect of pyrroloquinoline quinone (PQQ) on skin aging in the Bmi-1 KO mice and its underlying mechanisms, we administered a normal diet to both Wild type mice (WT) and Bmi-1 KO mice, while supplementing the diet of Bmi-1 KO mice with PQQ (PQQ+Bmi-1 KO). Subsequently, we compared the thickness of the skin epidermis, dermis, pilosebaceous unit and collagen ratio using HE staining and Masson's trichrome. Additionally, immunohistochemical staining, Western blotting and electron microscopy were applied across all three groups. The results revealed that Bmi-1 KO mice exhibited premature aging phenotypes compared to the WT group; however, PQQ administration effectively delayed premature aging in Bmi-1 KO mice. Furthermore, reduced epidermal thickness, dermal thickness, pilosebaceous units count as well as collagen ratio were observed in Bmi-1 KO mice. Moreover, the PCNA positive cell percentage also decreased in Bmi-1 KO mice. Conversely, treatment with PQQ significantly increased epidermal thickness, dermal thickness, pilosebaceous unit count, collagen ratio and PCNA positive cell percentage when compared to Bmi-1 KO mice. In order to further investigate the anti-aging mechanism of PQQ, experiments have revealed that PQQ effectively suppressed the expression of cell cycle proteins p16, p19, and p53 in Bmi-1 KO mice. In addition, autophagy-related experiments demonstrated that compared to the WT group, Bmi-1 KO mice exhibited an increased number of autophagosomes along with decreased expression of Beclin-1 and LC3Ⅱ/LC3Ⅰratio, and increased expression of p62. However, supplementation with PQQ resulted in a reduction in the number of autophagosomes while increasing the expression of Beclin-1 and LC3Ⅱ/LC3Ⅰratio and decreasing the expression of p62. This study provides evidence that downregulation of Bmi-1 promotes skin aging, whereas PQQ delays skin aging in Bmi-1 KO mice by promoting cell proliferation, inhibiting the expression of p16, p19 and p53 and enhancing autophagy levels.
Collapse
Affiliation(s)
- Bin Li
- Department of Stomatology, Hunan University of Medicine, Huaihua, China
| | - Xiao Meng Yang
- Department of Sonography, Hunan University of Medicine General Hospital, Huaihua China
| | - Xiong Ming Zhou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuan Qing Huang
- Department of Stomatology, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
10
|
Xia B, Liang J, Lu Y, Ding J, Peng J, Li F, Dai J, Liu Y, Wang J, Wan C, Luo P. Lactoferrin influences atherosclerotic progression by modulating macrophagic AMPK/mTOR signaling-dependent autophagy. Sci Rep 2025; 15:10585. [PMID: 40148507 DOI: 10.1038/s41598-025-95181-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
This study aimed to explore the role of lactoferrin (LTF) in atherosclerosis (AS) and its possible mechanisms. Human left coronary artery tissues were collected and divided into control (CON), coronary heart disease (CHD) and sudden coronary death (SCD) groups. Pathologic changes (including changes in the coronary plaque area, necrotic core, collagen fibers, and foam cell content) were observed. The LTF, P62, and 4-hydroxynonenal (4-HNE) expression levels were assessed. The ApoE-/- AS mouse model was established. The pathological changes and related protein levels were analyzed after autophagy inhibition. The foam cell model was constructed using an ox-LDL-induced human monocyte line, THP-1. The LTF, BECN1, LC3-II/I, AMP-activated protein kinase (AMPK)/the mammalian target of rapamycin (mTOR) pathway proteins, B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), and 4-HNE expressions were then detected after silencing of LTF or BECN1. Plaque stability was significantly lower in the SCD group compared to the non-SCD group (p < 0.05). LTF, P62 and 4-HNE levels in plaques increased as plaque stability decreased, and LTF was significantly correlated with plaque progression and autophagy levels. Autophagy inhibition by U0126 leads to the worsening of aortic luminal stenosis, increased necrotic core and foam cell deposits, decreased autophagosomes, reduced LTF expression, and upregulated P62 expression in AS mice. It was further demonstrated that LTF expression correlates with autophagy. LTF expression was increased in ox-LDL-treated THP-1 cells, and silencing BECN1 and/or LTF increased mTOR phosphorylation and 4-HNE levels, inhibited BECN1 and LC3 II expression and AMPK activation, and simultaneously decreased the Bcl-2/Bax ratio. LTF might alleviate AS pathology through accelerating the AMPK/mTOR pathway, and suggested that LTF may be a potential predictive molecule for AS.
Collapse
Affiliation(s)
- Bing Xia
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China.
| | - Jingwei Liang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yanlin Lu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jin Peng
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Fangqin Li
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jialin Dai
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Yubo Liu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jie Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Changwu Wan
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Peng Luo
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
11
|
Huang X, Yan H, Xu Z, Yang B, Luo P, He Q. The inducible role of autophagy in cell death: emerging evidence and future perspectives. Cell Commun Signal 2025; 23:151. [PMID: 40140912 PMCID: PMC11948861 DOI: 10.1186/s12964-025-02135-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Autophagy is a lysosome-dependent degradation pathway for recycling intracellular materials and removing damaged organelles, and it is usually considered a prosurvival process in response to stress stimuli. However, increasing evidence suggests that autophagy can also drive cell death in a context-dependent manner. The bulk degradation of cell contents and the accumulation of autophagosomes are recognized as the mechanisms of cell death induced by autophagy alone. However, autophagy can also drive other forms of regulated cell death (RCD) whose mechanisms are not related to excessive autophagic vacuolization. Notably, few reviews address studies on the transformation from autophagy to RCD, and the underlying molecular mechanisms are still vague. AIM OF REVIEW This review aims to summarize the existing studies on autophagy-mediated RCD, to elucidate the mechanism by which autophagy initiates RCD, and to comprehensively understand the role of autophagy in determining cell fate. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the prodeath effect of autophagy, which is distinct from the generally perceived cytoprotective role, and its mechanisms are mainly associated with the selective degradation of proteins or organelles essential for cell survival and the direct involvement of the autophagy machinery in cell death. Additionally, this review highlights the need for better manipulation of autophagy activation or inhibition in different pathological contexts, depending on clinical purpose.
Collapse
Affiliation(s)
- Xiangliang Huang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China.
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, China.
| |
Collapse
|
12
|
Lu JJ, Ning Y, Hu WT, Sheng YR, Liu YK, Xie F, Li MQ, Zhu XY. Excess heme orchestrates progesterone resistance in uterine endometrial cancer through macrophage polarization and the IL-33/PAX8/PGR axis. Biomed Pharmacother 2025; 186:118008. [PMID: 40138919 DOI: 10.1016/j.biopha.2025.118008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Progesterone is an important drug for hormone therapy in uterine endometrial cancer (UEC). However, the therapeutic efficacy of progestogen is often limited by resistance, and the underlying mechanism remains unknown. In this study, we observed heme metabolism is more active in progesterone-insensitive patients. Heme induced macrophages (Mφs) bias towards M2-like phenotype and downregulated the expression of IL-33, resulting in increased levels of Paired box gene 8 (PAX8). Further study showed PAX8 inhibited the transcriptional activity of PGR by binding to the PGR promoter region. In addition, PGR can also act as a transcriptional factor to regulate the transcription of autophagy-related gene 7 (ATG). Low expression of PGR decreases the transcriptional activity of ATG7 promoter, which decreases cell autophagy and promotes the progression of UEC. Overall, this study reveals the important interaction between heme metabolism, IL-33 and PGR in progesterone-insensitive UEC, and is promising to provide new therapeutic targets for overcoming progesterone resistance.
Collapse
Affiliation(s)
- Jia-Jing Lu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, PR China; Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China
| | - Yan Ning
- Department of Pathology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China
| | - Wen-Ting Hu
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China
| | - Yan-Ran Sheng
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China
| | - Yu-Kai Liu
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China
| | - Feng Xie
- Medical Center of Diagnosis and Treatment for Cervical and Intrauterine Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, PR China
| | - Ming-Qing Li
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China; Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| | - Xiao-Yong Zhu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, PR China; Department of Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, PR China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, PR China.
| |
Collapse
|
13
|
Luong TV, Cao MTT, Nguyen NVD, Dang HNN, Nguyen TT. Roles of autophagy and long non-coding RNAs in gastric cancer. World J Gastroenterol 2025; 31:101124. [PMID: 40124267 PMCID: PMC11924004 DOI: 10.3748/wjg.v31.i11.101124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 03/13/2025] Open
Abstract
Gastric cancer (GC) is one of the most aggressive malignancies worldwide and is characterized by its poor prognosis and resistance to conventional therapies. Autophagy and long non-coding RNAs (lncRNAs) play critical yet complex roles in GC, functioning as both tumor suppressors and promoters depending on the disease stage and context. Autophagy influences cellular homeostasis and metabolism, whereas lncRNAs regulate gene expression through epigenetic modifications, RNA sponging, and protein interactions. Notably, the interplay between lncRNAs and autophagy modulates tumor progression, metastasis, chemoresistance, and the tumor microenvironment. This study explored the intricate relationship between lncRNAs and autophagy in GC, highlighting their roles in pathogenesis and treatment resistance. By addressing current knowledge gaps and proposing innovative therapeutic strategies, we have emphasized the potential of targeting this dynamic interplay for improved diagnostic and therapeutic outcomes.
Collapse
Affiliation(s)
- Thang Viet Luong
- Department of Internal Medicine, University of Medicine and Pharmacy, Hue University, Hue 530000, Viet Nam
| | - Mai Thi Thu Cao
- Department of Biochemistry, University of Medicine and Pharmacy, Hue University, Hue 530000, Viet Nam
| | - Nam Van Duc Nguyen
- Department of Internal Medicine, University of Medicine and Pharmacy, Hue University, Hue 530000, Viet Nam
| | | | - Trung Tran Nguyen
- Department of Biotechnology, NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Viet Nam
| |
Collapse
|
14
|
Wei M, Wu Y, Yang Q, Zhou Z, Xu X. Serum Starvation Regulates Autophagy of Human Periodontal Ligament Cells Through Reactive Oxygen Species Mediated Adenosine Monophosphate-Activated Protein Kinase/Mechanistic Target of RAPAMYCIN Axis. Int Dent J 2025; 75:1461-1471. [PMID: 40120460 DOI: 10.1016/j.identj.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/03/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
INTRODUCTION AND AIMS Human periodontal ligament cells (hPDLCs) play a pivotal role in periodontal tissue remodelling, a process essential for orthodontic tooth movement (OTM). Autophagy, a survival mechanism under cellular stress, is induced by nutrient deprivation and impacts hPDLC function. This study aimed to explore the role of autophagy in the adaptive response of hPDLCs to nutritional stress, an environment simulating conditions during OTM. METHODS Nutrient deprivation in hPDLCs was modelled through serum starvation. Autophagy levels and relevant markers were assessed using electron microscopy, protein assays, and gene expression analyses. Emphasis was placed on adenosine monophosphate-activated protein kinase (AMPK) signalling, specifically phosphorylation of AMPKα at Thr172, as a regulatory node in autophagy induction. Loss- and gain-of-function approaches were utilized to investigate the role of Thr172 in AMPK-mediated autophagy under nutrient stress. RESULTS Findings indicated a marked increase in reactive oxygen species-mediated autophagy in hPDLCs under nutrient deprivation. This process was significantly regulated by AMPK activation through Thr172 phosphorylation, establishing AMPK as a critical factor in autophagy induction during cellular adaptation to nutritional stress. CONCLUSION Nutritional stress enhances reactive oxygen species-mediated autophagy in hPDLCs via AMPK signalling, underscoring the role of autophagy in cellular adaptation during OTM. Targeting the AMPK pathway could provide novel insights for optimizing orthodontic treatment by leveraging cellular adaptive mechanisms. CLINICAL RELEVANCE Understanding the molecular mechanisms underlying autophagy in hPDLCs opens potential therapeutic pathways to improve OTM outcomes. Modulating autophagy may lead to advances in orthodontic therapies that facilitate periodontal tissue remodelling, enhancing clinical effectiveness.
Collapse
Affiliation(s)
- Mianxing Wei
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Yujie Wu
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Qian Yang
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Zheng Zhou
- University of Detroit Mercy, School of Dentistry, Graduate Periodontics, Detroit, Michigan, USA.
| | - Xiaomei Xu
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China.
| |
Collapse
|
15
|
Ponce-Mora A, Salazar NA, Domenech-Bendaña A, Locascio A, Bejarano E, Gimeno-Mallench L. Interplay Between Polyphenols and Autophagy: Insights From an Aging Perspective. FRONT BIOSCI-LANDMRK 2025; 30:25728. [PMID: 40152368 DOI: 10.31083/fbl25728] [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: 07/16/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 03/29/2025]
Abstract
The relationship between polyphenols and autophagy, particularly in the context of aging, presents a promising avenue for therapeutic interventions in age-related diseases. A decline in autophagy is associated with aging-related affections, and an increasing number of studies suggest that this enhancement is linked to cellular resilience and longevity. This review delves into the multifaceted roles of autophagy in cellular homeostasis and the potential of polyphenols to modulate autophagic pathways. We revised the most updated literature regarding the modulatory effects of polyphenols on autophagy in cardiovascular, liver, and kidney diseases, highlighting their therapeutic potential. We highlight the role of polyphenols as modulators of autophagy to combat age-related diseases, thus contributing to improving the quality of life in aging populations. A better understanding of the interplay of autophagy between autophagy and polyphenols will help pave the way for future research and clinical applications in the field of longevity medicine.
Collapse
Affiliation(s)
- Alejandro Ponce-Mora
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
| | - Nicolle Andrea Salazar
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
| | - Alicia Domenech-Bendaña
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
| | - Antonella Locascio
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
| | - Eloy Bejarano
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
| | - Lucia Gimeno-Mallench
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
| |
Collapse
|
16
|
Milano G, Reinero M, Puyal J, Tozzi P, Samaja M, Porte-Thomé F, Beghetti M. Inhibition of Sodium/Hydrogen Exchanger-1 in the Right Ventricle and Lung Dysfunction Induced by Experimental Pulmonary Arterial Hypertension in Rats. J Am Heart Assoc 2025; 14:e036859. [PMID: 40055146 DOI: 10.1161/jaha.124.036859] [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: 08/15/2024] [Accepted: 11/06/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Life-threatening pulmonary arterial hypertension (PAH) still lacks a direct therapeutic approach targeted to the molecular defects associated with the disease. Here, we focus on the impaired regulation of intracellular acidity and sodium/calcium overload by testing the hypothesis that inhibiting NHE-1 (sodium/hydrogen exchanger isoform 1) with rimeporide enables the recovery of pulmonary and right ventricular dysfunctions in the Sugen5416/hypoxia PAH model in rats. METHODS AND RESULTS Adult Sprague-Dawley male rats (n=44) rats were divided into 2 broad groups: control and Sugen5416/hypoxia. After verifying PAH insurgence in the Sugen5416/hypoxia group by transthoracic echocardiography and pulse-wave Doppler analysis, rats were treated with either 100 mg/kg per day rimeporide or placebo in drinking water for 3 weeks. The functional, morphological (fibrosis and hypertrophy), and biochemical (inflammation, signaling pathways) dysfunctions caused by PAH were partially reverted by rimeporide in both the lungs and myocardium, where the most striking effects were observed in the right ventricle. Rimeporide improved hemodynamics in the pulmonary circulation and in the right ventricle, with decrease in right ventricle hypertrophy, pulmonary vascular remodeling, inflammation, and fibrosis. No effect of rimeporide was detected in control rats. The protective effect of rimeporide was accompanied by decreased p-Akt/Akt (phosphorylated protein kinase B/protein kinase B) ratio and increased autophagy flux mainly in the right ventricle. CONCLUSIONS By specifically inhibiting NHE-1, rimeporide at the selected dosage revealed remarkable anti-PAH effects by preventing the functional, morphological, and biochemical deleterious effects of PAH on the right ventricle and lungs. Rimeporide should be considered as a potential treatment for PAH.
Collapse
MESH Headings
- Animals
- Rats, Sprague-Dawley
- Male
- Sodium-Hydrogen Exchanger 1/metabolism
- Sodium-Hydrogen Exchanger 1/antagonists & inhibitors
- Disease Models, Animal
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/prevention & control
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Lung/drug effects
- Lung/metabolism
- Lung/physiopathology
- Ventricular Function, Right/drug effects
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/prevention & control
- Hypertrophy, Right Ventricular/etiology
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/prevention & control
- Pyrroles/pharmacology
- Pulmonary Artery/drug effects
- Pulmonary Artery/physiopathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats
- Signal Transduction/drug effects
- Heart Ventricles/physiopathology
- Heart Ventricles/metabolism
- Heart Ventricles/drug effects
- Heart Ventricles/pathology
- Indoles
Collapse
Affiliation(s)
- Giuseppina Milano
- Department Cœur-Vaisseaux, Cardiac Surgery Center University Hospital of Lausanne Switzerland
| | - Melanie Reinero
- Department Cœur-Vaisseaux, Cardiac Surgery Center University Hospital of Lausanne Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences University of Lausanne Switzerland
- CURML, University Center of Legal Medicine, Lausanne University Hospital Lausanne Switzerland
| | - Piergiorgio Tozzi
- Department Cœur-Vaisseaux, Cardiac Surgery Center University Hospital of Lausanne Switzerland
| | | | | | - Maurice Beghetti
- Unité de Cardiologie Pédiatrique University Hospital of Geneva, University of Geneva Switzerland
| |
Collapse
|
17
|
Xue D, Huang J, Sun X, Zhang W, Ma H, Yin D, Wang Y, Wang J, Yang C, Geng Q. Dissection of the potential mechanism of polystyrene microplastic exposure on cardiomyocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179048. [PMID: 40101404 DOI: 10.1016/j.scitotenv.2025.179048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Microplastics (MPs) are ubiquitous in the global biosphere, have widespread contact with humans, and increase exposure risks. Increasing evidence indicates that MPs exposure increases the risks of cardiovascular disease, however, a comprehensive exploration of the fundamental cellular mechanisms has yet to be undertaken. In this study, we used AC16 cells as a model and exposed them to 10 to 50 μg/mL of polystyrene MPs (PS-MPs), chosen based on the average daily intake and absorption of MPs by humans, to investigate their roles and mechanisms in cell injury. Proteomic analysis reveals that PS-MP-induced differentially expressed genes were enriched on endoplasmic reticulum (ER) stress and autophagy-related entries. The findings from immunofluorescence and western blotting provided further verification of the activation of ER stress by PS-MPs. Although the expression of LC3-II, a canonical autophagy marker was increased, PS-MPs inhibited autophagic flux instead of inducing autophagy. Importantly, ER stress not only contributes to PS-MPs-induced cell injury but also involved in PS-MPs-induced autophagic flux inhibition. Furthermore, the inhibition of autophagy, and the partial restoration of cell injury induced by PS-MPs was achieved through the activation of autophagy. Overall, the results reveal that activation of ER stress and inhibition of autophagic flux plays a significant role in the cell injury caused by PS-MPs in human cardiomyocytes, offering a novel perspective on the mechanism behind MPs-induced cardiomyocyte toxicity.
Collapse
Affiliation(s)
- Dahui Xue
- Department of Geriatrics, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
| | - Jingnan Huang
- Department of Geriatrics, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
| | - Xin Sun
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
| | - Wei Zhang
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Huan Ma
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou, 510000, China
| | - Da Yin
- Department of Cardiology, Shenzhen Cardiovascular Minimally Invasive Medical Engineering Technology Research and Development Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
| | - Yuanhao Wang
- Department of Geriatrics, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
| | - Jigang Wang
- Department of Geriatrics, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China; Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.; State Key Laboratory for Quality Esurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Chuanbin Yang
- Department of Geriatrics, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China.
| | - Qingshan Geng
- Department of Geriatrics, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China.
| |
Collapse
|
18
|
Pourhabib Mamaghani M, Mousavikia SN, Azimian H. Ferroptosis in cancer: Mechanisms, therapeutic strategies, and clinical implications. Pathol Res Pract 2025; 269:155907. [PMID: 40101548 DOI: 10.1016/j.prp.2025.155907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/10/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
The resistance of cancer cells to existing treatments has become a major challenge for researchers despite advancements in cancer treatment. Studies have shown that this resistance is due to cancer cells evading apoptosis. Moreover, the most common form of cell death induced by chemotherapy and radiotherapy is apoptosis. One of the most essential mechanisms cancer cells escape apoptosis is the excessive expression of tumors' apoptosis inhibitors. Therefore, finding a non-apoptotic pathway that bypasses apoptosis could be a hopeful strategy for cancer treatment. Ferroptosis has been identified as a non-apoptotic and regulated cell death process characterized by the accumulation of lipid peroxides and iron-dependent reactive oxygen species (ROS). Although studies have shown that ferroptosis plays a role in the development of many diseases, including cancer, it also has the potential to decrease resistance to current treatments, such as chemotherapy. Additionally, research has shown that ferroptosis successfully kills cancer cells, such as breast, stem, and lung cancer cells. Therefore, ferroptosis can be identified as a beneficial therapeutic mechanism for cancer treatment. Although ferroptosis has been introduced as an effective treatment path for cancer, its role, along with its therapeutic inducers, in increasing the therapeutic effect has not been investigated. In this review, we aim to introduce ferroptosis, compare it with other cell deaths known so far, and explain its role in cancer treatment. We believe that ferroptosis can be widely used to overcome cancer cells.
Collapse
Affiliation(s)
- Mina Pourhabib Mamaghani
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nasibeh Mousavikia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Wang W, Li T, Wu K. Cell death in tumor microenvironment: an insight for exploiting novel therapeutic approaches. Cell Death Discov 2025; 11:93. [PMID: 40064873 PMCID: PMC11894105 DOI: 10.1038/s41420-025-02376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cell death is critical in tumor biology. The common cancer therapies can cause cell death and alleviate tumor, while the cancer cells can develop a resistance to cell death and survive from the therapies. Thus, not only observing the alternative mechanisms of tumor cells resistant to cell death, but also understanding the intricate dynamics of cell death processes within the tumor microenvironment (TME), are essential for tailoring effective therapeutic strategies. High-throughput sequencing technologies have revolutionized cancer research by enabling comprehensive molecular profiling. Recent advances in single cell sequencing have unraveled the heterogeneity of TME components, shedding light on their complex interactions. In this review, we explored the interplay between cell death signaling and the TME, summarised the potential drugs inducing cell death in pre-clinical stage, reviewed some studies applying next-generation sequencing technologies in cancer death research, and discussed the future utilization of updated sequencing platforms in screening novel treatment methods targeted cell death. In conclusion, leveraging multi-omics technologies to dissect cell death signaling in the context of the TME holds great promise for advancing cancer research and therapy development.
Collapse
Affiliation(s)
- Wenxin Wang
- BGI Genomics, Shenzhen, 518083, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310030, China
| | - Tong Li
- BGI Genomics, Shenzhen, 518083, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310030, China
| | - Kui Wu
- BGI Genomics, Shenzhen, 518083, China.
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI Research, Shenzhen, 518083, China.
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), BGI Research, Hangzhou, 310030, China.
| |
Collapse
|
20
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
21
|
Zhang Y, Chen Y, Wu C, Cai Z, Yao W, Yang H, Song J, Xie X, Zhang L, Yi C. Establishment of a yeast essential protein conditional-degradation library and screening for autophagy-regulating genes. Autophagy 2025:1-13. [PMID: 39988731 DOI: 10.1080/15548627.2025.2469189] [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: 07/15/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved intracellular degradation pathway that relies on vacuoles or lysosomes. Over 40 ATG genes have been identified in yeast cells as participants in various types of autophagy, although these genes are non-essential. While some essential genes involved in autophagy have been identified using temperature-sensitive yeast strains, systematic research on essential genes in autophagy remains lacking. To address this, we established an essential protein conditional degradation library using the auxin-inducible degron (AID) system. By introducing the GFP-Atg8 plasmid, we identified 29 essential yeast genes involved in autophagy, 19 of which had not been previously recognized. In summary, the yeast essential protein conditional degradation library we constructed will serve as a valuable resource for systematically investigating the roles of essential genes in autophagy and other biological functions.Abbreviation: AID: auxin-inducible degron; ALP: alkaline phosphatase; ATG: autophagy related; CSG: constitutive slow growth; DAmP: Decreased Abundance by mRNA Perturbation; GFP: green fluorescent protein; MMS: methyl methanesulfonate; ORF: open reading frame; PAS: phagophore assembly site; PCR: polymerase chain reaction; SD-G: glucose starvation medium; SD-N: nitrogen starvation medium; TOR: target of rapamycin kinase; YGRC: yeast genetic resource center; YPD: yeast extract peptone dextrose.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yingcong Chen
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Choufei Wu
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Zhengyi Cai
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Weijing Yao
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huan Yang
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Song
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Xiankuan Xie
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University school of Medicine, Hangzhou, Zhejiang, China
| | - Liqin Zhang
- Biology Department, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Life Sciences, Huzhou University, Huzhou, Zhejiang, China
| | - Cong Yi
- Department of Biochemistry and Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Li B, Liu J, Zhang D, Chu Y, Chen Z, Tsao J, Chen T, Jiang J, Hu K. Evodiamine Promotes Autophagy and Alleviates Oxidative Stress in Dry Eye Disease Through the p53/mTOR Pathway. Invest Ophthalmol Vis Sci 2025; 66:44. [PMID: 40111353 PMCID: PMC11932426 DOI: 10.1167/iovs.66.3.44] [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: 10/22/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Purpose This study aims to explore the therapeutic efficacy of evodiamine (EVO) in the treatment of dry eye disease (DED). Methods Mouse models of DED was developed using benzalkonium chloride eye drops and subcutaneous atropine injections. Corneal epithelial defects were assessed by fluorescein sodium staining, and tear secretion was measured with the phenol red thread test. For the in vitro model, human corneal epithelial cells were cultured in a sodium chloride-enriched medium. Phenotypic and mechanistic analyses were conducted using real-time quantitative PCR, Western blotting, flow cytometry, and immunofluorescence staining. Results The administration of EVO eye drops significantly enhanced tear secretion in mice, ameliorated ocular surface damage, decreased the expression of corneal inflammatory factors, and increased the density of conjunctival goblet cells. Furthermore, EVO reduced oxidative stress by promoting autophagy. Mechanistically, EVO-induced autophagy was mediated via the p53/mammalian target of rapamycin pathway. Conclusions These findings suggest that EVO is a potential therapeutic agent for the treatment of DED, with its beneficial effects attributed to the activation of autophagy through the p53/mammalian target of rapamycin pathway.
Collapse
Affiliation(s)
- Boda Li
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Junpeng Liu
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Di Zhang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiran Chu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zeying Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiaruei Tsao
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Taige Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
23
|
Wang Y, Pei W, Yang Y, Xia C, Zhang Q, Geng Z, Shi X, Wang F. Inhibition of XIST restrains paclitaxel resistance in breast cancer cells by targeting hsa-let-7d-5p/ATG16L1 through regulation of autophagy. Cell Signal 2025; 127:111534. [PMID: 39638138 DOI: 10.1016/j.cellsig.2024.111534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer is a fatal malignant tumor in women worldwide. The development of paclitaxel resistance remains a challenge. Autophagy is considered to have a significant part in the chemotherapeutic stress mechanism. This study aimed to investigate the function of long non-coding RNA (lncRNA) in breast cancer cell chemoresistance and autophagy. The paclitaxel (PTX)-resistant breast cancer cells were established. The function of X-inactive specific transcript (XIST) was demonstrated using in vitro and in vivo experiments. Transmission electron microscope (TEM) was used to observe autophagy vesicles. Protein and mRNA levels were determined using western blotting and quantitative real time polymerase chain reaction (qRT-PCR). We discovered that autophagic activity was correlated with chemoresistance in PTX-resistant breast cancer cells. In vitro and in vivo studies showed that XIST inhibition reduced cell resistance to paclitaxel, caused autophagy to be suppressed by regulating hsa-let-7d-5p and ATG16L1 expression. Mechanically, threonine protein kinase B (PKB; also known as AKT) - mammalian target of rapamycin (mTOR) pathway was activated when knockdown of XIST, while was reversed by inhibition of hsa-let-7d-5p. Our results verified that XIST played a significant role in developing chemoresistance via mediating autophagy in PTX-resistant breast cancer cells. It may be a potential target for breast cancer treatment strategies.
Collapse
Affiliation(s)
- Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu 233004, Anhui, China; The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, Anhui, China
| | - Wenhao Pei
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Yuping Yang
- Department of Clinical Laboratory, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Chaoqun Xia
- Department of Clinical Laboratory, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Qiang Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu 233004, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Xiuru Shi
- Department of Blood Transfusion, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Fengchao Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China.
| |
Collapse
|
24
|
Chen Y, Li M, Wu Y. Heat shock protein 22: A new direction for cardiovascular disease (Review). Mol Med Rep 2025; 31:82. [PMID: 39886946 PMCID: PMC11800183 DOI: 10.3892/mmr.2025.13447] [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: 10/07/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
Small heat shock proteins (sHSPs) are common molecular chaperone proteins that function in various biological processes, and serve indispensable roles in maintaining cellular protein homeostasis and regulating the hydrolysis of unfolded proteins. HSP22 is a member of the sHSP family that is primarily expressed in the heart and skeletal muscle, as well as in various types of cancer. There have been important findings concerning the role of HSP22 in cardiovascular diseases. The aim of the present study was to provide insights into the various molecular mechanisms by which HSP22 functions in the heart, including oxidative stress, autophagy, apoptosis, the subcellular distribution of proteins and the promoting effect of proteasomes. In addition, drugs and cytokines, including geranylgeranylacetone, can exert protective effects on the heart by regulating the expression of HSP22. Based on increasingly abundant research, HSP22 may be considered a potential therapeutic target in cardiovascular diseases.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
25
|
Motiño O, Lambertucci F, Joseph A, Durand S, Anagnostopoulos G, Li S, Carbonnier V, Nogueira-Recalde U, Montégut L, Chen H, Aprahamian F, Nirmalathasan N, Maiuri MC, Pietrocola F, Valla D, Laouénan C, Gautier JF, Castera L, Martins I, Kroemer G. ACBP/DBI neutralization for the experimental treatment of fatty liver disease. Cell Death Differ 2025; 32:434-446. [PMID: 39550516 PMCID: PMC11894144 DOI: 10.1038/s41418-024-01410-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
Acyl-CoA binding protein (ACBP), also known as diazepam-binding inhibitor (DBI), is an extracellular checkpoint of autophagy. Here, we report that patients with histologically confirmed metabolic-associated steatohepatitis (MASH) or liver fibrosis exhibit elevated levels of circulating ACBP/DBI protein as compared to non-affected controls. Plasma ACBP/DBI strongly correlated with the NAFLD and FIB4 scores in patients, and these correlations were independent of age and body mass index. We studied the capacity of a monoclonal antibody (mAb) neutralizing mouse ACBP/DBI to combat active liver disease in several mouse models, in which steatohepatitis had been induced by four different protocols, namely, (i) methionine/choline-deficient diet, (ii) Western style diet (WD) alone, (iii) WD combined with the hepatotoxic agent CCl4, and (iv) a combination of CCl4 injections and oral ethanol challenge. Injections of anti-ACBP/DBI mAb attenuated histological, enzymological, metabolomic and transcriptomic signs of liver damage in these four models, hence halting or reducing the progression of non-alcoholic and alcoholic liver disease. Steatosis, inflammation, ballooning and fibrosis responded to ACBP/DBI inhibition at the preclinical level. Altogether, these findings support a causal role of ACBP/DBI in MASH and liver fibrosis, as well as the possibility to therapeutically target ACBP/DBI.
Collapse
Grants
- O.M is supported by the “Beatriz Galindo Junior” Program from the Spanish Ministry of Universities (BG22/00104).
- UN-R is supported by “Axudas de apoio á etapa de formación posdoutoral da Xunta de Galicia” – GAIN. N°Expediente: IN606B-2021/015.
- DL, CL, J-FG and LC were supported by RHU QUID‐NASH funded by Agence Nationale de la Recherche programe Investissements d'Avenir (Reference ANR- 17-T171105J-RHUS-0009) implemented by Institut National de la Recherche Medicale, Université Paris Cité, Centre de l'Energie Atomique, Labortaoires Servier, Biopredictive, and Assistance Publique-Hôpitaux de Paris.
- GK is supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR) – Projets blancs; Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; Fondation pour la Recherche Médicale (FRM); a donation by Elior; European Joint Programme on Rare Diseases (EJPRD) Wilsonmed; European Research Council Advanced Investigator Award (ERC-2021-ADG, Grant No. 101052444; project acronym: ICD-Cancer, project title: Immunogenic cell death (ICD) in the cancer-immune dialogue), The ERA4 Health Cardinoff Grant Ener-LIGHT, European Union Horizon 2020 research and innovation programmes Oncobiome (grant agreement number: 825410, Project Acronym: ONCOBIOME, Project title: Gut OncoMicrobiome Signatures [GOMS] associated with cancer incidence, prognosis and prediction of treatment response, Prevalung (grant agreement number 101095604, Project Acronym: PREVALUNG EU, project title: Biomarkers affecting the transition from cardiovascular disease to lung cancer: towards stratified interception), Neutrocure (grant agreement number 861878 : Project Acronym: Neutrocure ; project title: Development of “smart” amplifiers of reactive oxygen species specific to aberrant polymorphonuclear neutrophils for treatment of inflammatory and autoimmune diseases, cancer and myeloablation); National support managed by the Agence Nationale de la Recherche under the France 2030 programme (reference number 21-ESRE-0028, ESR/Equipex+ Onco-Pheno-Screen); Hevolution Network on Senescence in Aging; Institut National du Cancer (INCa); Institut Universitaire de France; LabEx Immuno-Oncology ANR-18-IDEX-0001; a Cancer Research ASPIRE Award from the Mark Foundation; PAIR-Obésité INCa_1873, the RHUs Immunolife and LUCA-pi (both dedicated to France Relance 2030); Seerave Foundation; SIRIC Cancer Research and Personalized Medicine (CARPEM). This study contributes to the IdEx Université de Paris Cité ANR-18-IDEX-0001.
Collapse
Affiliation(s)
- Omar Motiño
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Unidad de Excelencia, Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - CSIC, Valladolid, Spain.
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Gerasimos Anagnostopoulos
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sijing Li
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Uxía Nogueira-Recalde
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Profesor Novoa Santos, A Coruña, Spain
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Fanny Aprahamian
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Nitharsshini Nirmalathasan
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Naples, Italy
| | - Federico Pietrocola
- Department of Bioscience and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Dominique Valla
- Université Paris Cité, UMR1149 (CRI), Inserm, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Cédric Laouénan
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm U1137, Laboratory "Infection, Antimicrobials, Modelling, Evolution" (IAME), Paris, France
- Département d'Epidémiologie Biostatistique et Recherche Clinique, AP-HP.Nord, Hôpital Bichat, Paris, France
| | - Jean-François Gautier
- Institut Necker Enfants Malades, Inserm U1151, CNRS UMR 8253, IMMEDIAB Laboratory, Paris, France
- Centre Universitaire de Diabétologie et de ses Complications, AP-HP, Hôpital Lariboisiére, Paris, France
| | - Laurent Castera
- Université Paris Cité, UMR1149 (CRI), Inserm, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Institut Universitaire de France, Paris, France.
| |
Collapse
|
26
|
Xian Y, Liu J, Dai M, Zhang W, He M, Wei Z, Jiang Y, Le S, Lin Z, Tang S, Zhou Y, Dong L, Liang J, Zhang J, Wang L. Microglia Promote Lymphangiogenesis Around the Spinal Cord Through VEGF-C/VEGFR3-Dependent Autophagy and Polarization After Acute Spinal Cord Injury. Mol Neurobiol 2025; 62:2740-2755. [PMID: 39158788 DOI: 10.1007/s12035-024-04437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Reducing secondary injury is a key focus in the field of spinal cord injury (SCI). Recent studies have revealed the role of lymphangiogenesis in reducing secondary damage to central nerve. However, the mechanism of lymphangiogenesis is not yet clear. Macrophages have been shown to play an important role in peripheral tissue lymphangiogenesis. Microglia is believed to play a role similar to macrophages in the central nervous system (CNS); we hypothesized that there was a close relationship between microglia and central nerve system lymphangiogenesis. Herein, we used an in vivo model of SCI to explored the relationship between microglia and spinal cord lymphangiogenesis and further investigated the polarization of microglia and its role in promoting spinal cord lymphangiogenesis by a series of in vitro experiments. The current study elucidated for the first time the relationship between microglia and lymphangiogenesis around the spinal cord after SCI. Classical activated (M1) microglia can promote lymphangiogenesis by secreting VEGF-C which further increases polarization and secretion of lymphatic growth factor by activating VEGFR3. The VEGF-C/VEGFR3 pathway activation downregulates microglia autophagy, thereby regulating the microglia phenotype. These results indicate that M1 microglia promote lymphangiogenesis after SCI, and activated VEGF-C/VEGFR3 signaling promotes M1 microglia polarization by inhibiting autophagy, thereby facilitates lymphangiogenesis.
Collapse
Affiliation(s)
- Yeyang Xian
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Liu
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Mengxuan Dai
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Wensheng Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Minye He
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhengnong Wei
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yutao Jiang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shiyong Le
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhuoang Lin
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shuai Tang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yunfei Zhou
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Liming Dong
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jinzheng Liang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| | - Liang Wang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| |
Collapse
|
27
|
Liang N, Cao Y, Li J, Zhang K. Normal dermal mesenchymal stem cells improve the functions of psoriatic keratinocytes by inducing autophagy. Acta Histochem 2025; 127:152229. [PMID: 39864345 DOI: 10.1016/j.acthis.2025.152229] [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: 05/27/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVE Psoriasis is a chronic inflammatory skin disease characterized by excessive proliferation and abnormal differentiation of keratinocytes. Although stem cell-based therapies have shown promise in treating psoriasis, the underlying mechanisms remain unclear. This study aimed to established a psoriatic cell model to investigate the effect of normal dermal mesenchymal stem cell (DMSCs) on keratinocyte proliferation, inflammation responses and the associated mechanism. METHODS To create an in vitro model of psoriasis, HaCaT cells were stimulated with a mixture of five inflammatory cytokines including IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α (M5). A transwell co-culture system was employed to assess the influence of normal DMSCs on proliferation and inflammation response of HaCaT cells. Cell viability was assessed using the CCK-8 assay and EDU incorporation assay. The expression levels of mRNA for inflammatory cytokines (IL-8, IL-17A and TNF-α) in HaCaT cells co-cultured with either normal or psoriatic DMSCs were quantified by qRT-PCR. Apoptosis was evaluated by annexin V-FITC/PI double staining and TUNEL/DAPI staining assay. Autophagy was detected by immunostaining, RT-PCR and western blotting. Additionally, the expression levels of mRNA and protein of both Akt and mammalin target of rapamycin(mTOR) were also determined. RESULTS Normal DMSCs were found to decrease the viability and promote apoptosis of HaCaT cells treated with M5. Furthermore, DMSCs reduced the secretion of proinflammatory cytokines, such as IL-8, IL-17A and TNF-α. Importantly, normal DMSCs were shown to induced autophagy in HaCaT cell. Pretreatment of HaCaT cells with autophagy inhibitor 3-methyladenine (3-MA) reversed the anti-psoriatic effect of normal DMSCs. Notably, DMSCs promote autophagy in M5-treated HaCaT cells by inhibition of p-Akt/Akt and p-mTOR/mTOR ratio. CONCLUSION Normal mesenchymal stem cells promote autophagy through the inhibition of Akt/mTOR signaling pathway, leading to the alleviation of psoriasis in vitro. These findings provide insights into the potential mechanisms by which DMSCs may exert therapeutic effects in psoriasis and support further investigation into their clinical applications.
Collapse
Affiliation(s)
- Nannan Liang
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yue Cao
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cell for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
28
|
Lu D, Wu S, Wang X, Zhang J, Xu Y, Tao L, Shen X. Oxymatrine alleviates ALD-induced cardiac hypertrophy by regulating autophagy via activation Nrf2/SIRT3 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156389. [PMID: 39827815 DOI: 10.1016/j.phymed.2025.156389] [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: 07/30/2024] [Revised: 12/24/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Cardiac hypertrophy is a prevalent early pathological manifestation in various cardiovascular diseases, lacking effective interventions to impede its progression. Although oxymatrine (OMT) has shown potential benefits for cardiac function, its therapeutic efficacy and mechanism in cardiac hypertrophy remain incompletely understood. Notably, mitochondrial damage and dysregulated autophagy are pivotal pathogenic mechanisms in cardiac hypertrophy. PURPOSE We investigate the pharmacological characteristics and mechanism of OMT in mitochondrial function and autophagy in cardiac hypertrophy. STUDY DESIGN AND METHODS A murine model of cardiac hypertrophy was induced by aldosterone in combination with high-salt drinking water, while primary cardiomyocyte hypertrophy was induced by aldosterone in vitro. Cardiac hypertrophy was assessed using echocardiography and histopathological staining. Autophagosomes and mitochondrial morphology were visualized by transmission electron microscopy. Levels of reactive oxygen species (ROS), malondialdehyde (MDA), and adenosine triphosphate (ATP) were quantified using commercial kits. The binding affinity of OMT with Nrf2 was assessed through molecular docking. Furthermore, adenovirus, agonists, and inhibitors were employed to modulate Nrf2, followed by quantitative real-time polymerase chain reaction (qRT-PCR), immunoblotting, co-immunoprecipitation, chromatin immunoprecipitation, immunohistochemistry, and cellular thermal shift assay. RESULTS OMT effectively attenuated aldosterone-induced cardiac hypertrophy both in vivo and in vitro. OMT promoted the activation of Nrf2, leading to elevated SIRT3 expression and enhanced autophagolysosome fusion, thereby modulating mitophagy and improving mitochondrial function. Moreover, the cardioprotective effects of OMT were abolished upon silencing or inhibition of Nrf2. OMT binds to Nrf2, facilitating its dissociation and nuclear translocation. CONCLUSION OMT activates Nrf2, consequently enhancing SIRT3 transcription, restoring autophagic flux, and preserving mitochondrial integrity, thereby mitigating aldosterone-induced cardiac hypertrophy. In summary, our study is the first to discover and confirm that OMT can stabilize Nrf2, promoting its activation and subsequently up-regulating SIRT3, which in turn facilitates mitochondrial autophagy. Additionally, PARKIN appears to play a key role in SIRT3-mediated regulation of mitophagy, warranting further investigation.
Collapse
Affiliation(s)
- Dingchun Lu
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China
| | - Shun Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China
| | - Xueting Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China
| | - Jian Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China
| | - Yini Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China; The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang City and Guian New District, Guizhou 561113, China.
| |
Collapse
|
29
|
Chen Z, Zheng N, Wang F, Zhou Q, Chen Z, Xie L, Sun Q, Li L, Li B. The role of ferritinophagy and ferroptosis in Alzheimer's disease. Brain Res 2025; 1850:149340. [PMID: 39586368 DOI: 10.1016/j.brainres.2024.149340] [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: 07/02/2024] [Revised: 10/17/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
Iron is a crucial mineral element within human cells, serving as a pivotal cofactor for diverse biological enzymes. Ferritin plays a crucial role in maintaining iron homeostasis within the body through its ability to sequester and release iron. Ferritinophagy is a selective autophagic process in cells that specifically facilitates the degradation of ferritin and subsequent release of free iron, thereby regulating intracellular iron homeostasis. The nuclear receptor coactivator 4 (NCOA4) serves as a pivotal regulator in the entire process of ferritinophagy, facilitating its binding to ferritin and subsequent delivering to lysosomes for degradation, thereby enabling the release of free iron. The free iron ions within the cell undergo catalysis through the Fenton reaction, resulting in a substantial generation of reactive oxygen species (ROS). This process induces lipid peroxidation, thereby stimulating a cascade leading to cellular tissue damage and subsequent initiation of ferroptosis. Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive deterioration of emotional memory and cognitive function, accompanied by mental and behavioral aberrations. The pathology of the disease is characterized by aberrant deposition of amyloid β-protein (Aβ) and hyperphosphorylated tau protein. It has been observed that evident iron metabolism disorders and accumulation of lipid peroxides occur in AD, indicating a significant impact of ferritinophagy and ferroptosis on the pathogenesis and progression of AD. This article elucidates the process and mechanism of ferritinophagy and ferroptosis, investigating their implications in AD to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ziwen Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Nan Zheng
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Fuwei Wang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Qiong Zhou
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zihao Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Lihua Xie
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Qiang Sun
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Li Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| | - Baohong Li
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The Affiliated Dongguan Songshan Lake Central Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
30
|
Xiao N, Liu H, Zhang R, Li Y, Zhao X, Chen H, Zhang C, Zhu Y, Lu H, Wang X, Liu H, Wan J. N-acetyltransferase 10 impedes EZH2/H3K27me3/GABARAP axis mediated autophagy and facilitates lung cancer tumorigenesis through enhancing SGK2 mRNA acetylation. Int J Biol Macromol 2025; 297:139823. [PMID: 39814292 DOI: 10.1016/j.ijbiomac.2025.139823] [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: 11/23/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
N4-acetylcytidine (ac4C) is a critical RNA modification implicated in cancer progression. Currently, N-acetyltransferase 10 (NAT10) is recognized as the sole "writer" protein responsible for ac4C modification. However, the study of NAT10 and ac4C modification in lung cancer remains sparse. In this study, we observed a significant upregulation of NAT10 expression in lung cancer, which is strongly correlated with poor prognostic outcomes. In vitro and in vivo experiments have demonstrated that NAT10 facilitates the proliferation, migration, and invasion of non-small cell lung cancer (NSCLC) cells while inhibiting autophagy flux. Mechanistically, NAT10 may enhance mRNA stability through ac4c modification at the 3' untranslated region (UTR) of SGK2 mRNA. Furthermore, SGK2 interacts with EZH2 and phosphorylates it at threonine 367, leading to increased protein stability of EZH2 and a reduction in its ubiquitination. Additionally, NAT10 impedes autophagy flux by preventing the fusion of autophagosomes with lysosomes and suppressing GABARAP transcription, which is regulated by EZH2-mediated H3K27me3. In summary, our study elucidates the biological significance and molecular mechanisms of the NAT10/SGK2/EZH2 axis in the pathogenesis of lung cancer, potentially providing novel prognostic markers and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruike Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangzhuan Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenxing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongshen Lu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuanzhi Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
31
|
Dabravolski SA, Churov AV, Beloyartsev DF, Kovyanova TI, Lyapina IN, Sukhorukov VN, Orekhov AN. The role of NRF2 function and regulation in atherosclerosis: an update. Mol Cell Biochem 2025:10.1007/s11010-025-05233-y. [PMID: 40025257 DOI: 10.1007/s11010-025-05233-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025]
Abstract
Atherosclerosis, a chronic inflammatory disease of the arteries, remains a leading cause of cardiovascular morbidity and mortality worldwide. This review examines the molecular mechanisms underlying NRF2 role in atherosclerosis, focusing on the recently defined intricate interplay between autophagy, the nuclear factor erythroid 2-related factor 2 (NRF2) pathway, microRNAs (miRNAs), and genes regulating NRF2 with atheroprotective effects. The NRF2/autophagy axis emerges as a critical regulator of cellular responses to oxidative stress and inflammation in atherosclerosis, with key players including Heat Shock Protein 90 (HSP90), Neuropeptide Y (NPY), and Glutaredoxin 2 (GLRX2). MiRNAs are identified as potent regulators of gene expression in atherosclerosis, impacting NRF2 signalling and disease susceptibility. Additionally, genes such as Prenyl diphosphate synthase subunit 2 (PDSS2), Sulfiredoxin1 (Srxn1), and Isocitrate dehydrogenase 1 (IDH1) are implicated in NRF2-dependent atheroprotective pathways. Future research directions include elucidating the complex interactions between these molecular pathways, evaluating novel therapeutic targets in preclinical and clinical settings, and addressing challenges related to drug delivery and patient heterogeneity. Despite limitations, this review underscores the potential for targeted interventions aimed at modulating NRF2/autophagy signalling and miRNA regulatory networks to mitigate atherosclerosis progression and improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, 2161002, Karmiel, Israel.
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A. V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Tatiana I Kovyanova
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Irina N Lyapina
- Research Institute for Complex Issues of Cardiovascular Diseases, 6 Barbarash Boulevard, Kemerovo, Russia, 650002
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| |
Collapse
|
32
|
Rivagorda M, Romeo-Guitart D, Blanchet V, Mailliet F, Boitez V, Barry N, Milunov D, Siopi E, Goudin N, Moriceau S, Guerrera C, Leibovici M, Saha S, Codogno P, Morselli E, Morel E, Armand AS, Oury F. A primary cilia-autophagy axis in hippocampal neurons is essential to maintain cognitive resilience. NATURE AGING 2025; 5:450-467. [PMID: 39984747 PMCID: PMC11922775 DOI: 10.1038/s43587-024-00791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/06/2024] [Indexed: 02/23/2025]
Abstract
Blood-borne factors are essential to maintain neuronal synaptic plasticity and cognitive resilience throughout life. One such factor is osteocalcin (OCN), a hormone produced by osteoblasts that influences multiple physiological processes, including hippocampal neuronal homeostasis. However, the mechanism through which this blood-borne factor communicates with neurons remains unclear. Here we show the importance of a core primary cilium (PC) protein-autophagy axis in mediating the effects of OCN. We found that the OCN receptor GPR158 is present at the PC of hippocampal neurons and mediates the regulation of autophagy machinery by OCN. During aging, autophagy and PC core proteins are reduced in neurons, and restoring their levels is sufficient to improve cognitive impairments in aged mice. Mechanistically, the induction of this axis by OCN is dependent on the PC-dependent cAMP response element-binding protein signaling pathway. Altogether, this study demonstrates that the PC-autophagy axis is a gateway to mediate communication between blood-borne factors and neurons, and it advances understanding of the mechanisms involved in age-related cognitive decline.
Collapse
Affiliation(s)
- Manon Rivagorda
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - David Romeo-Guitart
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Victoria Blanchet
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - François Mailliet
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Valérie Boitez
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Natalie Barry
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | | | - Eleni Siopi
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Nicolas Goudin
- Platform for Image Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Stéphanie Moriceau
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
- Platform for Neurobehavioral and Metaboblism, Institut Imagine, Structure Fédérative de Recherche Necker, 26 INSERM US24/CNRS UAR, Paris, France
| | - Chiara Guerrera
- Platform for Proteomic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Michel Leibovici
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | | | - Patrice Codogno
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 6, Paris, France
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Etienne Morel
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 6, Paris, France
| | - Anne-Sophie Armand
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Franck Oury
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France.
| |
Collapse
|
33
|
ALMatrafi TA. Deciphering the role of TMEM164 in autophagy-mediated ferroptosis and immune modulation in non-small cell lung cancer. Cell Immunol 2025; 409-410:104915. [PMID: 39798196 DOI: 10.1016/j.cellimm.2024.104915] [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: 08/04/2024] [Revised: 11/13/2024] [Accepted: 12/25/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains one of the most prevalent and deadly malignancies. Despite advancements in molecular therapies and diagnostic methods, the 5-year survival rate for lung adenocarcinoma patients remains unacceptably low, highlighting the urgent need for novel therapeutic strategies. Ferroptosis, a distinct form of regulated cell death, has emerged as a promising target in cancer treatment. This study investigates the role of TMEM164, a membrane protein, in promoting ferroptosis and modulating anti-tumor immunity in NSCLC, aiming to elucidate its therapeutic potential. METHODS Using publicly available datasets, we performed bioinformatics analyses to identify TMEM164-regulated genes involved in ferroptosis. In addition, in vitro and in vivo assays were conducted to assess the impact of TMEM164 on cellular functions in NSCLC. RESULTS Functional assays demonstrated that TMEM164 overexpression significantly inhibited invasion, migration, and cell proliferation in both in vitro and in vivo models. TMEM164 was also found to induce ferroptosis in NSCLC cells by promoting autophagy. Specifically, we identified a mechanism whereby TMEM164 mediates ATG5-dependent autophagosome formation, leading to the degradation of ferritin, GPX4, and lipid droplets. This degradation facilitated iron accumulation and lipid peroxidation, which triggered iron-dependent cell death. Notably, co-administration of TMEM164 upregulation and anti-PD-1 antibodies exhibited synergistic anti-tumor effects in a mouse model. CONCLUSION These findings suggest that targeting TMEM164 to enhance ferroptosis and stimulate anti-tumor immunity may inhibit NSCLC progression. Consequently, TMEM164 holds promise as a new therapeutic target for NSCLC treatment.
Collapse
|
34
|
Cooper KF. Cargo hitchhiking autophagy - a hybrid autophagy pathway utilized in yeast. Autophagy 2025; 21:500-512. [PMID: 39757721 PMCID: PMC11849947 DOI: 10.1080/15548627.2024.2447207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/07/2025] Open
Abstract
Macroautophagy is a catabolic process that maintains cellular homeostasis by recycling intracellular material through the use of double-membrane vesicles called autophagosomes. In turn, autophagosomes fuse with vacuoles (in yeast and plants) or lysosomes (in metazoans), where resident hydrolases degrade the cargo. Given the conservation of autophagy, Saccharomyces cerevisiae is a valuable model organism for deciphering molecular details that define macroautophagy pathways. In yeast, macroautophagic pathways fall into two subclasses: selective and nonselective (bulk) autophagy. Bulk autophagy is predominantly upregulated following TORC1 inhibition, triggered by nutrient stress, and degrades superfluous random cytosolic proteins and organelles. In contrast, selective autophagy pathways maintain cellular homeostasis when TORC1 is active by degrading damaged organelles and dysfunctional proteins. Here, selective autophagy receptors mediate cargo delivery to the vacuole. Now, two groups have discovered a new hybrid autophagy mechanism, coined cargo hitchhiking autophagy (CHA), that uses autophagic receptor proteins to deliver selected cargo to phagophores built in response to nutrient stress for the random destruction of cytosolic contents. In CHA, various autophagic receptors link their cargos to lipidated Atg8, located on growing phagophores. In addition, the sorting nexin heterodimer Snx4-Atg20 assists in the degradation of cargo during CHA, possibly by aiding the delivery of cytoplasmic cargos to phagophores and/or by delaying the closure of expanding phagophores. This review will outline this new mechanism, also known as Snx4-assisted autophagy, that degrades an assortment of cargos in yeast, including transcription factors, glycogen, and a subset of ribosomal proteins.
Collapse
Affiliation(s)
- Katrina F. Cooper
- Department of Cell and Molecular Biology, Virtua Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| |
Collapse
|
35
|
Xu Z, Liu R, Ke H, Xu F, Yang P, Zhang W, Zhan Y, Zhao Z, Xiao F. ATP6V1D drives hepatocellular carcinoma stemness and progression via both lysosome acidification-dependent and -independent mechanisms. Autophagy 2025; 21:513-529. [PMID: 39316516 PMCID: PMC11849949 DOI: 10.1080/15548627.2024.2406186] [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: 05/15/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024] Open
Abstract
Metabolic reprogramming is pivotal in cancer stem cell (CSC) self-renewal. However, the intricate regulatory mechanisms governing the crosstalk between metabolic reprogramming and liver CSCs remain elusive. Here, using a metabolic CRISPR-Cas9 knockout screen, we identify ATP6V1D, a subunit of the vacuolar-type H+-translocating ATPase (V-ATPase), as a key metabolic regulator of hepatocellular carcinoma (HCC) stemness. Elevated ATP6V1D expression correlates with poor clinical outcomes in HCC patients. ATP6V1D knockdown inhibits HCC stemness and malignant progression both in vitro and in vivo. Mechanistically, ATP6V1D enhances HCC stemness and progression by maintaining macroautophagic/autophagic flux. Specifically, ATP6V1D not only promotes lysosomal acidification, but also enhances the interaction between CHMP4B and IST1 to foster ESCRT-III complex assembly, thereby facilitating autophagosome-lysosome fusion to maintain autophagic flux. Moreover, silencing CHMP4B or IST1 attenuates HCC stemness and progression. Notably, low-dose bafilomycin A1 targeting the V-ATPase complex shows promise as a potential therapeutic strategy for HCC. In conclusion, our study highlights the critical role of ATP6V1D in driving HCC stemness and progression via the autophagy-lysosomal pathway, providing novel therapeutic targets and approaches for HCC treatment.Abbreviations: 3-MA: 3-methyladenine; ANT: adjacent normal liver tissues; ATP6V1D: ATPase H+ transporting V1 subunit D; BafA1: bafilomycin A1; CHMP: charged multivesicular body protein; co-IP: co-immunoprecipitation; CSC: cancer stem cell; ESCRT: endosomal sorting complex required for transport; HCC: hepatocellular carcinoma; IF: immunofluorescence; IHC: immunohistochemical; LCSCs: liver cancer stem cells; qRT-PCR: quantitative real time PCR; V-ATPase: vacuolar-type H+- translocating ATPase; WB: western blot.
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Ruiyang Liu
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
| | - Haoying Ke
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
| | - Fuyuan Xu
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
| | - Pengfei Yang
- Department of Pathology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Weiyu Zhang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Yi Zhan
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
| | - Zhiju Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fei Xiao
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
- The Fifth Affiliated Hospital, Guangdong-Hong Kong-Macao University Joint of Interventional Medicine, Zhuhai, Guangdong Province, China
- State Key Laboratory of Anti-Infective Drug Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Kashi Guangdong Institute of Science and Technology, The First People’s Hospital of Kashi, Kashi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
36
|
Yi YS. Functional interplay between non-canonical inflammasomes and autophagy in inflammatory responses and diseases. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:129-138. [PMID: 39539180 PMCID: PMC11842290 DOI: 10.4196/kjpp.24.240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024]
Abstract
The inflammasome is a cytosolic multiprotein platform that plays a key role in the inflammatory response, an essential innate immune response that protects the body from pathogens and cellular danger signals. Autophagy is a fundamental cellular mechanism that maintains homeostasis through the elimination and recycling of dysfunctional molecules and subcellular elements. Many previous studies have demonstrated a functional interplay between canonical inflammasomes that were earlier discovered and autophagy in inflammatory responses and diseases. Given the increasing evidence that non-canonical inflammasomes are unique and key factors in inflammatory responses, the functional interplay between non-canonical inflammasomes and autophagy is noteworthy. Recent studies have demonstrated that non-canonical inflammasomes and autophagy are functionally correlated with inflammatory responses and diseases. This review comprehensively discusses recent studies that have investigated the functional interplay of non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4, with autophagy and autophagy-related proteins in inflammatory responses and diseases and provides insight into the development of novel anti-inflammatory therapeutics by modulating the functional interplay between non-canonical inflammasomes and autophagy.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Korea
| |
Collapse
|
37
|
Ding Y, Jing W, Kang Z, Yang Z. Exploring the role and application of mitochondria in radiation therapy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167623. [PMID: 39674289 DOI: 10.1016/j.bbadis.2024.167623] [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: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Mitochondria are pivotal in cellular energy metabolism, the oxidative stress response and apoptosis. Recent research has focused on harnessing their functions to enhance the efficacy of radiation therapy (RT). This review focuses on the critical functions and applications of mitochondria in radiation therapy, including the targeting of mitochondrial metabolism and the modulation of mitochondria-mediated cell death and immune responses. While these strategies have demonstrated considerable potential in preclinical studies to improve radiotherapy outcomes, challenges remain, such as optimizing drug delivery systems, ensuring safety and overcoming resistance to therapy.
Collapse
Affiliation(s)
- Yi Ding
- Shandong University, Jinan 250000, China
| | - Wang Jing
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Zhichao Kang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Zhe Yang
- Shandong University, Jinan 250000, China.
| |
Collapse
|
38
|
Li S, Mingoia S, Montégut L, Lambertucci F, Chen H, Dong Y, De Palma FDE, Scuderi SA, Rong Y, Carbonnier V, Martins I, Maiuri MC, Kroemer G. Atlas of expression of acyl CoA binding protein/diazepam binding inhibitor (ACBP/DBI) in human and mouse. Cell Death Dis 2025; 16:134. [PMID: 40011442 DOI: 10.1038/s41419-025-07447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Acyl CoA binding protein encoded by diazepam binding inhibitor (ACBP/DBI) is a tissue hormone that stimulates lipo-anabolic responses and inhibits autophagy, thus contributing to aging and age-related diseases. Protein expression profiling of ACBP/DBI was performed on mouse tissues to identify organs in which this major tissue hormone is expressed. Transcriptomic and proteomic data bases corroborated a high level of human-mouse interspecies conservation of ACBP/DBI expression in different organs. Single-cell RNA-seq data confirmed that ACBP/DBI was strongly expressed by parenchymatous cells from specific human and mouse organs (e.g., kidney, large intestine, liver, lung) as well as by myeloid or glial cells from other organs (e.g., adipose tissue, brain, eye) following a pattern that was conserved among the two species. We identified a panel of 44 mRNAs that are strongly co-expressed with ACBP/DBI mRNA in normal and malignant human and normal mouse tissues. Of note, 22 (50%) of these co-expressed mRNAs encode proteins localized at mitochondria, and mRNAs with metabolism-related functions are strongly overrepresented (66%). Systematic data mining was performed to identify transcription factors that regulate ACBP/DBI expression in human and mouse. Several transcription factors, including growth response 1 (EGR1), E2F Transcription Factor 1 (E2F1, which interacts with retinoblastoma, RB) and transformation-related protein 53 (TRP53, best known as p53), which are endowed with oncosuppressive effects, consistently repress ACBP/DBI expression as well as its co-expressed mRNAs across multiple datasets, suggesting a mechanistic basis for a coregulation network. Furthermore, we identified multiple transcription factors that transactivate ACBP/DBI gene expression together with its coregulation network. Altogether, this study indicates the existence of conserved mechanisms determining the expression of ACBP/DBI in specific cell types of the mammalian organism.
Collapse
Affiliation(s)
- Sijing Li
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Silvia Mingoia
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Pharmacological Sciences, University of Piemonte Orientale, Novara, Italy
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Yanbing Dong
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Fatima Domenica Elisa De Palma
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Sarah Adriana Scuderi
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Yan Rong
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
39
|
Cheng CY, Chen KP, Tseng TS, Hua KF, Ju TC. The therapeutic potential of (R)-carvedilol in Huntington's disease through enhancement of autophagy-lysosomal pathway via GSK-3β inhibition. Neurotherapeutics 2025:e00557. [PMID: 40011132 DOI: 10.1016/j.neurot.2025.e00557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by the abnormal expansion of CAG repeats in the huntingtin (Htt) gene, leading to the aggregation of mutant huntingtin protein (mHTT) in cells, particularly in cortical and striatal neurons. This results in involuntary movements, cognitive impairment, and emotional instability. One of the critical pathogenic mechanisms in HD is impaired autophagy, which plays a vital role in cellular homeostasis by degrading damaged organelles and misfolded proteins through the formation of autophagosomes that fuse with lysosomes. However, the aggregation of mHTT disrupts autophagic function, leading to the accumulation of mHTT and exacerbating the disease's pathogenesis. Carvedilol is an established clinical medication used to treat hypertension and congestive heart failure. It exerts protective effects by blocking both β1-and β2-adrenergic receptors, reducing sympathetic nervous activity, and promoting vasodilation through α1-adrenergic blockade. Carvedilol has been shown to possess antioxidant and anti-inflammatory properties. In this study, we demonstrate that (R)-carvedilol promotes the nuclear translocation of the transcription factor binding to IGHM enhancer 3 (TFE3) by reducing glycogen synthase-3β (GSK-3β) activation, which increases the expression of autophagy-related proteins and facilitates the autophagy-lysosomal pathway (ALP), thereby enhancing mHTT degradation. Additionally, systemic administration of (R)-carvedilol improves mHTT degradation, provides neuroprotection, and inhibits gliosis, effectively ameliorating behavioral impairments and improving disease progression. Overall, these findings indicate that (R)-carvedilol has therapeutic potential for managing HD by promoting autophagy, facilitating the clearance of mHTT aggregates, and demonstrating advantageous properties in an HD transgenic mouse model, highlighting its promise as a treatment option for neurodegenerative diseases.
Collapse
Affiliation(s)
- Chih-Yuan Cheng
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407224, Taiwan; Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei 114201, Taiwan
| | - Kai-Po Chen
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407224, Taiwan
| | - Tien-Sheng Tseng
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402202, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260007, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| | - Tz-Chuen Ju
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402202, Taiwan; Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan.
| |
Collapse
|
40
|
Guo N, Xia Y, He N, Zhang L, Liu J. IRGM Inhibits the AKT/mTOR Signaling Pathway by Interacting with TRIM21 to Alleviate Sepsis-Induced Acute Lung Injury. Inflammation 2025:10.1007/s10753-025-02265-w. [PMID: 39994091 DOI: 10.1007/s10753-025-02265-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025]
Abstract
Acute lung injury (ALI) is a severe complication of sepsis, and its underlying pathological mechanisms remain poorly understood. This study aims to investigate the role and mechanisms by which IRGM mediates autophagy through the regulation of the AKT/mTOR signaling pathway in sepsis-induced ALI. Initially, a sepsis-induced ALI mouse model was established using cecal ligation and puncture (CLP). Our results demonstrated that Irgm1 expression was significantly upregulated in the ALI model. Subsequently, Irgm1 was knocked down in vivo using AAV vectors, and changes in ALI symptoms were assessed. In vitro, a sepsis-induced ALI cell model was generated by stimulating A549 cells with lipopolysaccharide (LPS). The effects of IRGM overexpression on autophagy and apoptosis were evaluated, and its impact on the AKT/mTOR signaling pathway was analyzed. Furthermore, mass spectrometry and co-immunoprecipitation (COIP) experiments were conducted to explore the interaction between IRGM and TRIM21. In vivo results showed that Irgm1 knockout exacerbated CLP-induced ALI, as evidenced by a significant reduction in autophagic activity, increased apoptosis, and aberrant activation of the AKT/mTOR pathway. Further cellular experiments suggested that IRGM may enhance autophagy by inhibiting the AKT/mTOR signaling pathway, thereby attenuating LPS-induced cell damage. Additionally, COIP experiments revealed that IRGM interacts with TRIM21 to inhibit AKT/mTOR pathway activation, thereby promoting autophagy and mitigating sepsis-induced ALI. In conclusion, IRGM regulates autophagy through the AKT/mTOR signaling pathway and exerts protective effects in sepsis-induced ALI, suggesting that it may serve as a potential therapeutic target for sepsis-related ALI.
Collapse
Affiliation(s)
- Na Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yu Xia
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China
| | - Nannan He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China
| | - Lei Zhang
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou, Gansu Province, China.
| | - Jian Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China.
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou, Gansu Province, China.
| |
Collapse
|
41
|
Liu W, Xia K, Huang X, Wei Z, Wei Z, Wang X, Xiong C, Guo W. HMGCL activates autophagy in osteosarcoma through β-HB mediated inhibition of the PI3K/AKT/mTOR signaling pathway. J Transl Med 2025; 23:219. [PMID: 39985081 PMCID: PMC11846287 DOI: 10.1186/s12967-025-06227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND 3-hydroxy-3-methylglutaryl-coenzymOHBe A(HMG-CoA) lyase (HMGCL) catalyzes the cleavage of HMG-CoA into acetyl-CoA and acetoacetic acid and serves as a rate-limiting enzyme in the metabolism of ketone bodies. While HMGCL is involved in various biological processes, its specific role in osteosarcoma remains unclear. METHODS Using data from a public database of osteosarcoma patients, we investigated the expression and prognostic value of HMGCL. The effects of HMGCL on the proliferation, migration, and invasion of osteosarcoma cells were assessed using CCK-8 assays, wound healing tests, and transwell invasion assays. We explored and validated the specific molecular mechanisms by which HMGCL influences osteosarcoma through transcriptome sequencing. Finally, we established a subcutaneous tumor formation model in nude mice to investigate the function of HMGCL in vivo. RESULTS The expression of HMGCL is downregulated in osteosarcoma and correlates with the prognosis of osteosarcoma patients. Overexpression of HMGCL can inhibit the proliferation, migration, and invasion of osteosarcoma cells, as well as tumor growth in vivo. Through our investigation of the underlying mechanism, we found that HMGCL may inhibit the activation of the PI3K/AKT/mTOR signaling pathway via its product, β-HB. This inhibition promotes the phosphorylation of ULK1, thereby facilitating autophagy in osteosarcoma cells and enhancing the malignancy of the disease. CONCLUSION HMGCL inhibits the activation of the PI3K/AKT/mTOR signaling pathway mediated by β-HB, thereby reducing the proliferation, migration, and invasion of osteosarcoma cells while promoting autophagy. HMGCL may represent a new target for the treatment of osteosarcoma, offering new hope for patients with this disease.
Collapse
Affiliation(s)
- Wenda Liu
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Kezhou Xia
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Xinghan Huang
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Zhun Wei
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Zicheng Wei
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Xingyu Wang
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Chen Xiong
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China
| | - Weichun Guo
- Department of Orthopaedics, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
42
|
Gao X, Xiong Y, Ma H, Zhou H, Liu W, Sun Q. Visualizing bulk autophagy in vivo by tagging endogenous LC3B. Autophagy 2025:1-17. [PMID: 39952286 DOI: 10.1080/15548627.2025.2457910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
Macroautophagy/autophagy plays a crucial role in maintaining cellular and organismal health, making the measurement of autophagy flux in vivo essential for its study. Current tools often depend on the overexpression of autophagy probes. In this study, we developed a knock-in mouse model, termed tfLC3-KI, by inserting a tandem fluorescent tag coding sequence into the native Map1lc3b gene locus. We found that tfLC3-KI mice exhibit optimal expression of mRFP-eGFP-LC3B, allowing for convenient measurement of autophagic structures and flux at single-cell resolution, both in vivo and in primary cell cultures. Additionally, we compared autophagy in neurons and glial cells across various brain regions between tfLC3-KI mice and CAG-tfLC3 mice, the latter overexpressing the probe under the strong CMV promoter. Finally, we used tfLC3-KI mice to map the spatial and temporal dynamics of basal autophagy activity in the reproductive system. Our findings highlight the value of the tfLC3-KI mouse model for investigating autophagy flux in vivo and demonstrate the feasibility of tagging endogenous proteins to visualize autophagic structures and flux in both bulk and selective autophagy research in vivo.Abbreviation: BafA1: bafilomycin A1; CQ: chloroquine; EBSS: Earle's balanced salt solution; Es: elongating spermatids; HPF: hippocampalformation; HY: hypothalamus; LCs: leydig cells; OLF: olfactory areas; PepA: pepstatin A; Rs: round spermatids; SCs: sertoli cells; Spc: spermatocytes; Spg: spermatogonia; tfLC3: tandem fluorescently tagged mRFP-eGFP-LC3; TH: thalamus.
Collapse
Affiliation(s)
- Xiukui Gao
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yue Xiong
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hangbin Ma
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Hao Zhou
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qiming Sun
- Department of Respiratory and Critical Care Medicine, Center for Metabolism Research, The Fourth Affiliated Hospital of Zhejiang University School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Cui Y, Cao X, Zhang Y, Fu C, Li D, Sun Y, Zhang Y, Xu T, Tsukamoto T, Cao D, Jiang J. Protein phosphatase 1 regulatory subunit 15 A (PPP1R15A) promoted the progression of gastric cancer by activating cell autophagy under energy stress. J Exp Clin Cancer Res 2025; 44:52. [PMID: 39948597 PMCID: PMC11823012 DOI: 10.1186/s13046-025-03320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Glucose metabolism plays a critical role in tumor progression. When glucose intake is insufficient and the tumor's growth rate exceeds its energy supply, tumor cells typically adapt and overcome the energy stress through compensatory mechanisms to maintain the survival of tumor cells, which may also be related to tumor recurrence or metastasis. METHODS Different concentrations of glucose were selected as the basis for the energy stress model of gastric cancer. Then CCK-8 and flow cytometry were used to detect its effects on cell proliferation, apoptosis, and cell cycle. Differentially expressed genes (DEGs) were screened by RNA sequencing and the regulated pathways were identified by gene set enrichment analysis. The regulatory relationship between the gene PPP1R15A and its transcription factor JUN was proved by ChIP-qPCR and dual-luciferase reporter assay. The gain and loss of function assays were conducted to examine the effects of PPP1R15A under energy stress in vivo and in vitro. Potential regulatory mechanisms of PPP1R15A were further analyzed through a combination of online databases, RNA sequencing, and metabolite sequencing. The regulation of PPP1R15A on cell autophagy under energy stress was detected by western blot, transmission electron microscope, mRFP-GFP-LC3 adenovirus and laser scanning confocal microscopy. RESULTS PPP1R15A and the transcription factor JUN were significantly upregulated by glucose deprivation (0 mM vs. 25 mM), JUN combined with the promoter of PPP1R15A and activated its expression. Both PPP1R15A and JUN were highly expressed in gastric cancer tissues and were independent risk factors for prognosis in the gastric cancer cohort. Overexpression of PPP1R15A promoted cell proliferation, inhibited apoptosis, and was involved in cell cycle arrest. Further RNA and metabolite sequencing suggested that PPP1R15A was associated with cell autophagy. In vitro experiments confirmed that both glucose deprivation and overexpression of PPP1R15A promoted the biosynthesis of autolysosome and autophagosome, and activated the cleavage of LC3 complex in gastric cancer cells. Moreover, PPP1R15A knockdown inhibited cell autophagy induced by glucose deprivation. CONCLUSIONS PPP1R15A sustained the survival of gastric cancer cells by regulating autophagy under energy stress to resist or adapt to harsh environments.
Collapse
Affiliation(s)
- Yingnan Cui
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Doctor of excellence program (DEP), The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yangyu Zhang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chenhao Fu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuzheng Zhang
- Department of Hospital Infection Management, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Tingshuang Xu
- Core facility of The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology I, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
44
|
Huang H, Liang X, Li S, Yan Y, Li S, Qiu C, Ye Z, Zhu Y, Shen D, Lin Y, Wang L, Chen N, Yao Y, Zhao X, Wu F, Shi X, Kou L, Chen R, Yao Q. Chondrocyte-targeted bilirubin/rapamycin carrier-free nanoparticles alleviate oxidative stress and modulate autophagy for osteoarthritis therapy. J Control Release 2025; 378:517-533. [PMID: 39701459 DOI: 10.1016/j.jconrel.2024.12.024] [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: 09/07/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic disease, characterized by the destruction of joint cartilage and synovitis, affects over 7 % of people worldwide. Disease-modifying treatments for OA still face significant challenges. Chondrocytes, as the exclusive cellular component of articular cartilage, play a pivotal role in synthesizing the intricate matrix of cartilage, thereby assuming a critical responsibility in facilitating its renewal and repair processes. However, oxidative stress within chondrocytes and subsequent apoptotic cell death plays significant roles in the progression of OA. Therefore, targeting apoptosis inhibition and mitigation of oxidative stress in chondrocytes represents a promising therapeutic strategy for OA. This study develops a type II collagen-targeting peptide (WYRGRLC) modified bilirubin/rapamycin carrier-free nanoparticle (PP/BRRP) and evaluate its therapeutic potential for OA. The PP/BRRP system exhibits remarkable chondrocyte-targeting ability, enabling the rupture of highly oxidized chondrocytes and subsequent release of bilirubin and rapamycin. This dual payload effectively scavenges reactive oxygen species, triggers autophagy, and suppresses the mTOR pathway, thereby augmenting anti-inflammatory and anti-apoptotic effects. The in vivo experiments further validate the retention and therapeutic efficacy of PP/BRRP in rat joints affected by OA. Overall, PP/BRRP exhibits significant potential for intervention and treatment of OA.
Collapse
Affiliation(s)
- Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xindan Liang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengjie Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yuqi Yan
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shize Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhanzheng Ye
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yixuan Zhu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Dingchao Shen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinhao Lin
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Luhui Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Nuo Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinsha Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Fugen Wu
- Department of Pediatric, The First People's Hospital of Wenling, Taizhou, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
45
|
Shen Y, Jiang K, Tan D, Zhu M, Qiu Y, Huang P, Zou W, Deng J, Wang Z, Xiong Y, Hong D. uN2CpolyG-mediated p65 nuclear sequestration suppresses the NF-κB-NLRP3 pathway in neuronal intranuclear inclusion disease. Cell Commun Signal 2025; 23:68. [PMID: 39920690 PMCID: PMC11806584 DOI: 10.1186/s12964-025-02079-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/01/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Neuronal intranuclear inclusion disease (NIID) is genetically linked to CGG repeat expansion in the 5'-untranslated region of the NOTCH2NLC gene, with nascent polyglycine-containing protein (uN2CpolyG) identified as a primary pathogenic factor. Emerging clinical evidence suggests that inflammation contributes to NIID pathogenesis, yet the underlying molecular mechanisms remain elusive. This study aimed to elucidate the molecular interaction between uN2CpolyG and the NF-κB-NLRP3 pathway. METHODS Single-cell RNA sequencing was conducted on the skin tissues of NIID patients to assess changes in the expression of genes involved in inflammatory pathways. Cell models (HEK-293T and U87-MG) transfected with CGG9/69/100 expansion vectors were used to investigate alterations in the NF-κB-NLRP3-autophagy pathway. Additionally, the therapeutic potential of NF-κB activators was evaluated in a Drosophila model with a CGG expansion knock-in. RESULTS Single-cell sequencing revealed a significant reduction in the expression of NFKBIA, encoding NF-κB inhibitor alpha (IkBa), which facilitates the nuclear translocation of p65, a key NF-κB component. uN2CpolyG directly interacted with and sequestered p65 in nuclear inclusions, leading to reduced phosphorylated p65 (p-p65) levels. This sequestration significantly downregulated the NF-κB-NLRP3 pathway, impairing autophagy, as indicated by decreased LC3II/LC3I ratios. Treatment of CGG100 cells with lipopolysaccharide (LPS) significantly increased p-p65, NLRP3, and LC3II/LC3I levels while reducing insoluble uN2CpolyG levels and intranuclear inclusions. In the Drosophila knock-in model, LPS significantly reduced the number of intranuclear inclusions and improved phenotypic manifestations. CONCLUSIONS This study revealed that uN2CpolyG directly interacts with and sequesters p65, thereby inhibiting the NF-κB-NLRP3 pathway and impairing autophagy. This mechanism highlights a novel therapeutic target for NIID and provides potentially broader insights into similar mechanisms in other neurodegenerative diseases characterized by misfolded protein aggregates.
Collapse
Affiliation(s)
- Yu Shen
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
| | - Kaiyan Jiang
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
| | - Dandan Tan
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China
- Rare Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Min Zhu
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China
- Rare Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yusen Qiu
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China
- Rare Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Pencheng Huang
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health Commission, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenquan Zou
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ying Xiong
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China.
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China.
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health Commission, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yong Wai Zheng Street 17#, Nanchang, 330006, China.
- Institute of Neurology, The First Affiliated Hospital, Jiangxi Medical College, Jiangxi Academy of Clinical Medical Science, Nanchang University, Nanchang, China.
- Rare Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health Commission, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
46
|
Cook ASI, Chen M, Nguyen TN, Cabezudo AC, Khuu G, Rao S, Garcia SN, Yang M, Iavarone AT, Ren X, Lazarou M, Hummer G, Hurley JH. Structural pathway for PI3-kinase regulation by VPS15 in autophagy. Science 2025:eadl3787. [PMID: 39913640 DOI: 10.1126/science.adl3787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 09/30/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
The class III phosphatidylinositol-3 kinase complexes I and II (PI3KC3-C1 and -C2) have vital roles in macroautophagy and endosomal maturation, respectively. We elucidated a structural pathway of enzyme activation through cryo-EM analysis of PI3KC3-C1. The inactive conformation of the VPS15 pseudokinase stabilizes the inactive conformation, sequestering its N-myristate in the N-lobe of the pseudokinase. Upon activation, the myristate is liberated such that the VPS34 lipid kinase catalyzes PI3P production on membranes. The VPS15 pseudokinase domain binds tightly to guanosine triphosphate (GTP), and stabilizes a web of interactions to autoinhibit the cytosolic complex and to promote the activation upon membrane binding. These findings show in atomistic detail how the VPS34 lipid kinase is activated in the context of a complete PI3K complex.
Collapse
Affiliation(s)
- Annan S I Cook
- Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Minghao Chen
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Thanh N Nguyen
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Ainara Claveras Cabezudo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- IMPRS on Cellular Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Grace Khuu
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Shanlin Rao
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- IMPRS on Cellular Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Samantha N Garcia
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Mingxuan Yang
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Anthony T Iavarone
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Xuefeng Ren
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Michael Lazarou
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Gerhard Hummer
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- IMPRS on Cellular Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Institute of Biophysics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - James H Hurley
- Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
47
|
Liu Y, He Y, Lei S. The important role of cuproptosis and cuproptosis-related genes in the development of thyroid carcinoma revealed by transcriptomic analysis and experiments. Braz J Otorhinolaryngol 2025; 91:101560. [PMID: 39914044 PMCID: PMC11848474 DOI: 10.1016/j.bjorl.2025.101560] [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: 07/30/2024] [Revised: 12/10/2024] [Accepted: 12/28/2024] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE Programmed Cell Death (PCD) processes have been suggested to play a role in the development of cancers. The study aimed to investigate the enrichment of different types of PCD and identify important PCD-related genes in Thyroid Carcinoma (THCA). METHODS The whole study is based on transcriptomic analysis and Immunohistochemistry (IHC) experiments. For transcriptomic analysis, the transcriptomic data of THCA (n = 493) and normal thyroid samples (n = 58) was used. The enrichment score of different types of PCD in THCA samples was calculated and the most enriched PCD process was identified. Then the expression of PCD-related genes in control and THCA samples were compared and the association between cuproptosis and characteristics of Tumor Microenvironment (TME) in THCA tissues was explored. IHC experiment was performed to confirm gene expressions in THCA and para-tumor samples. RESULTS Enrichment analysis showed that cuproptosis was the most enriched type of PCD process and associated with malignancy in THCA. The expression of a cuproptosis-related gene, CDKN2A, was found and confirmed to be higher in THCA than normal samples and associated with poor outcomes and higher clinical stages of patients with THCA. Moreover, CDKN2A expression was associated with immunosuppressive TME in THCA. CONCLUSION Our findings indicated the important role of cuproptosis and a cuproptosis-related gene, CDKN2A, in the development and progression of THCA, which might provide novel insights into the understanding of pathophysiology of THCA.
Collapse
Affiliation(s)
- Yani Liu
- Wuhan No. 1 Hospital, Department of Otolaryngology and Head and Neck Surgery, Wuhan, China
| | - Yanyan He
- Shengjing Hospital of China Medical University, Department of Otolaryngology and Head and Neck Surgery, Shenyang, China
| | - Shizhen Lei
- Wuhan No. 1 Hospital, Department of Ophthalmology, Wuhan, China.
| |
Collapse
|
48
|
Kim J, Lee Y, Jeon T, Ju S, Kim JS, Kim MS, Kang C. Autophagy-dependent splicing control directs translation toward inflammation during senescence. Dev Cell 2025; 60:364-378.e7. [PMID: 39510077 DOI: 10.1016/j.devcel.2024.10.008] [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: 04/18/2024] [Revised: 08/15/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024]
Abstract
The cellular proteome determines the functional state of cells and is often skewed to direct pathological conditions. Autophagy shapes cellular proteomes primarily through lysosomal degradation of either damaged or unnecessary proteins. Here, we show that autophagy directs the senescence-specific translatome to fuel inflammation by coupling selective protein degradation with alternative splicing. RNA splicing is significantly altered during senescence, some of which surprisingly depend on autophagy, including exon 5 skipping of the translation regulator EIF4H. Systematic translatome profiling indicates that this event is key to the translational bias toward inflammation in senescence. Autophagy promotes these changes by selectively degrading the splicing regulator splicing factor proline and glutamine rich (SFPQ) via the autophagy receptor NBR1. These autophagy-centric inflammatory controls appear to be conserved during human tissue aging and cancer. Our work highlights the role of autophagy in the on-demand functional remodeling of cellular proteomes as well as the crosstalk between autophagy, alternative splicing, and inflammatory translation.
Collapse
Affiliation(s)
- Jaejin Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for Systems Geroscience, Seoul National University, Seoul 08826, South Korea
| | - Yeonghyeon Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for Systems Geroscience, Seoul National University, Seoul 08826, South Korea
| | - Taerang Jeon
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for Systems Geroscience, Seoul National University, Seoul 08826, South Korea
| | - Seonmin Ju
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for RNA Research, Institute of Basic Science, Seoul 08826, South Korea
| | - Jong-Seo Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for RNA Research, Institute of Basic Science, Seoul 08826, South Korea
| | - Mi-Sung Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for Systems Geroscience, Seoul National University, Seoul 08826, South Korea
| | - Chanhee Kang
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for Systems Geroscience, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
49
|
Javed R, Mari M, Trosdal E, Duque T, Paddar MA, Allers L, Mudd MH, Claude-Taupin A, Akepati PR, Hendrix E, He Y, Salemi M, Phinney B, Uchiyama Y, Reggiori F, Deretic V. ATG9A facilitates the closure of mammalian autophagosomes. J Cell Biol 2025; 224:e202404047. [PMID: 39745851 PMCID: PMC11694768 DOI: 10.1083/jcb.202404047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/29/2024] [Accepted: 11/11/2024] [Indexed: 01/04/2025] Open
Abstract
Canonical autophagy captures within specialized double-membrane organelles, termed autophagosomes, an array of cytoplasmic components destined for lysosomal degradation. An autophagosome is completed when the growing phagophore undergoes ESCRT-dependent membrane closure, a prerequisite for its subsequent fusion with endolysosomal organelles and degradation of the sequestered cargo. ATG9A, a key integral membrane protein of the autophagy pathway, is best known for its role in the formation and expansion of phagophores. Here, we report a hitherto unappreciated function of mammalian ATG9A in directing autophagosome closure. ATG9A partners with IQGAP1 and key ESCRT-III component CHMP2A to facilitate this final stage in autophagosome formation. Thus, ATG9A is a central hub governing all major aspects of autophagosome membrane biogenesis, from phagophore formation to its closure, and is a unique ATG factor with progressive functionalities affecting the physiological outputs of autophagy.
Collapse
Affiliation(s)
- Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Muriel Mari
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Einar Trosdal
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Thabata Duque
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Masroor Ahmad Paddar
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Michal H. Mudd
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Aurore Claude-Taupin
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Prithvi Reddy Akepati
- Gastroenterology Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Emily Hendrix
- Department of Chemistry and Chemical Biology, The University of New Mexico, Albuquerque, NM, USA
| | - Yi He
- Department of Chemistry and Chemical Biology, The University of New Mexico, Albuquerque, NM, USA
| | - Michelle Salemi
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Brett Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
50
|
Li C, Yuan Y, Jia Y, Zhou Q, Wang Q, Jiang X. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies. Front Immunol 2025; 16:1534263. [PMID: 39963130 PMCID: PMC11830604 DOI: 10.3389/fimmu.2025.1534263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cellular aging is a multifactorial and intricately regulated physiological process with profound implications. The interaction between cellular senescence and cancer is complex and multifaceted, senescence can both promote and inhibit tumor progression through various mechanisms. M6A methylation modification regulates the aging process of cells and tissues by modulating senescence-related genes. In this review, we comprehensively discuss the characteristics of cellular senescence, the signaling pathways regulating senescence, the biomarkers of senescence, and the mechanisms of anti-senescence drugs. Notably, this review also delves into the complex interactions between senescence and cancer, emphasizing the dual role of the senescent microenvironment in tumor initiation, progression, and treatment. Finally, we thoroughly explore the function and mechanism of m6A methylation modification in cellular senescence, revealing its critical role in regulating gene expression and maintaining cellular homeostasis. In conclusion, this review provides a comprehensive perspective on the molecular mechanisms and biological significance of cellular senescence and offers new insights for the development of anti-senescence strategies.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Yixiao Yuan
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - YingDong Jia
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Zhou
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Xiulin Jiang
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|