1
|
Yuan Y, Kuang M, Yu T, Huang S, Jiang F, Lu B, Cai M, Lu X. Adipogenic dedifferentiation enhances survival of human umbilical cord-derived mesenchymal stem cells under oxidative stress. Adipocyte 2025; 14:2467150. [PMID: 39976240 PMCID: PMC11845070 DOI: 10.1080/21623945.2025.2467150] [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: 08/18/2024] [Revised: 12/16/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Mesenchymal stem cells (MSCs) serve as ideal candidates for a broad range of cell-based therapies. However, cell ageing caused by long-term in vitro expansion and poor survival after in vivo delivery greatly limits their success in preclinical and clinical applications. Dedifferentiation represents a potential strategy for enhancing the retention and function of MSCs in hostile environments. In this study, we evaluated the cell phenotype, proliferation, and differentiation potential, as well as the anti-oxidative stress ability of human umbilical cord-derived MSCs (hMSCs) manipulated with adipogenic priming and subsequent dedifferentiation. After an in vitro differentiation and dedifferentiation procedure, the resultant dedifferentiated hMSCs (De-hMSCs) displayed properties similar to their original counterparts, including immunophenotype and mesodermal potential. Upon re-induction, De-hMSCs exhibited a significantly higher adipogenic differentiation capability than unmanipulated hMSCs. Importantly, De-hMSCs showed a significantly enhanced ability to resist tert-butyl hydroperoxide (t-BHP) induced apoptosis compared to undifferentiated hMSCs. Mechanisms involving bcl-2 family proteins and autophagy may contribute to the demonstrated advantages of dedifferentiation-reprogrammed hMSCs. These results indicate that adipogenic dedifferentiation promotes adipogenesis and cell persistence, as well as preserves the stemness of human umbilical cord-derived MSCs that have been committed to the adipocytic lineage. As a unique stem cell population, dedifferentiated MSCs may represent an attractive and promising candidate for MSC-based therapy.
Collapse
Affiliation(s)
- Yin Yuan
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Meina Kuang
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tengye Yu
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Sirui Huang
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fujie Jiang
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Biyi Lu
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Mingen Cai
- School of Life Science & Biopharmacology, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin Lu
- School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
2
|
White B, Wang Z, Dean M, Michl J, Nieora N, Flannery S, Vendrell I, Fischer R, Hulikova A, Swietach P. Acidosis attenuates the hypoxic stabilization of HIF-1α by activating lysosomal degradation. J Cell Biol 2025; 224:e202409103. [PMID: 40552983 PMCID: PMC12187095 DOI: 10.1083/jcb.202409103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 04/12/2025] [Accepted: 05/19/2025] [Indexed: 06/28/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) mediate cellular responses to low oxygen, notably enhanced fermentation that acidifies poorly perfused tissues and may eventually become more damaging than adaptive. How pH feeds back on hypoxic signaling is unclear but critical to investigate because acidosis and hypoxia are mechanistically coupled in diffusion-limited settings, such as tumors. Here, we examined the pH sensitivity of hypoxic signaling in colorectal cancer cells that can survive acidosis. HIF-1α stabilization under acidotic hypoxia was transient, declining over 48 h. Proteomic analyses identified responses that followed HIF-1α, including canonical HIF targets (e.g., CA9, PDK1), but these did not reflect a proteome-wide downregulation. Enrichment analyses suggested a role for lysosomal degradation. Indeed, HIF-1α destabilization was blocked by inactivating lysosomes, but not proteasome inhibitors. Acidotic hypoxia stimulated lysosomal activity and autophagy via mammalian target of rapamycin complex I (mTORC1), resulting in HIF-1α degradation. This response protects cells from excessive acidification by unchecked fermentation. Thus, alkaline conditions are permissive for at least some aspects of HIF-1α signaling.
Collapse
Affiliation(s)
- Bobby White
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Zhenyi Wang
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew Dean
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Johanna Michl
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Natalia Nieora
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sarah Flannery
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Iolanda Vendrell
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Roman Fischer
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Xia F, Sha Y, Jin Y, Yang J, Chen C, Gong B, Liu Y, Zhao Q. Autophagy inhibition amplifies anti-tumor immunity effect of dinutuximab beta on neuroblastoma via the VEGFR/AKT/mTOR and ROS/NF-κB pathways. Int Immunopharmacol 2025; 158:114862. [PMID: 40378433 DOI: 10.1016/j.intimp.2025.114862] [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: 06/24/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Dinutuximab beta has shown limited efficacy in treating high-risk neuroblastoma (NB). Combining autophagy inhibitors with immune checkpoint inhibitors (ICIs) has proven effective in many malignancies. However, the anti-tumor effects of autophagy inhibition in conjunction with anti-GD2 immunotherapy remain unknown. In this study, dinutuximab beta induces anti-proliferation and anti-EMT activity in NB cells. Dinutuximab beta also triggers autophagy in NB cells, and inhibition of the VEGFR pathway with anlotinib amplifies dinutuximab beta-induced autophagy. In addition, dinutuximab beta induces the synthesis of the chemokine CXCL9 and the infiltration of CD8+ T cells. Mechanistically, dinutuximab beta inhibits the VEGFR/AKT/mTOR and ROS/NF-κB pathways. Furthermore, autophagy inhibition by CQ enhances CXCL9 expression and anti-tumor T cell responses of single anti-GD2 therapy in vitro and in vivo. Collectively, this study suggests autophagy inhibitors may be a promising strategy for enhancing therapeutic efficacy in NB in conjunction with anti-GD2 immunotherapy.
Collapse
Affiliation(s)
- Fantong Xia
- Radiation Oncology Center, Chongqing University Cancer Hospital, College of Medicine, Chongqing University, Chongqing, China
| | - Yongliang Sha
- Department of General Surgery, Xuzhou Central Hospital, Xuzhou, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
4
|
Shaykevich A, Chae D, Silverman I, Goel S, Maitra R. Modulating autophagy in KRAS mutant colorectal cancer using combination of oncolytic reovirus and carbamazepine. PLoS One 2025; 20:e0326029. [PMID: 40526688 DOI: 10.1371/journal.pone.0326029] [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: 02/17/2025] [Accepted: 05/20/2025] [Indexed: 06/19/2025] Open
Abstract
Oncolytic reovirus is a potential therapeutic for colorectal cancer patients with a mutant KRAS gene. Reovirus hijacks the autophagic machinery and preferentially induces apoptosis in patients with a KRAS mutation. However, reovirus on its own is not currently a viable treatment and requires enhancement with combination therapies. Carbamazepine, an FDA-approved drug in use for epilepsy, is an autophagy inducer and is used in this study in conjunction with reovirus. The dual treatment was able to reduce cancer viability in mutated KRAS cell lines and was more effective than with reovirus alone. Carbamazepine and reovirus increased autophagy-related proteins and mRNA in mutant KRAS compared to wildtype KRAS which is crucial for autophagy-induced apoptosis. Transmission electron microscopy results show increased autophagosome formation in the combination therapy, as well as a decrease in condensed chromatin. The combination therapy effectively increased the apoptosis induced by reovirus alone and is a viable treatment for patients with a mutant KRAS.
Collapse
Affiliation(s)
- Aaron Shaykevich
- Department of Biology, Yeshiva University, New York, New York, United States of America
| | - Danbee Chae
- Department of Biology, Yeshiva University, New York, New York, United States of America
| | - Isaac Silverman
- Department of Biology, Yeshiva University, New York, New York, United States of America
| | - Sanjay Goel
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, United States of America
| | - Radhashree Maitra
- Department of Biology, Yeshiva University, New York, New York, United States of America
| |
Collapse
|
5
|
Lyu Q, Kouketsu Y, Tazaki A, Kato M, Motooka Y, Toyokuni S. Terrestrial iron sulfide minerals induce distinct regulation of intracellular redox homeostasis and iron assimilation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118327. [PMID: 40381394 DOI: 10.1016/j.ecoenv.2025.118327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/12/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Repeated exposure to airborne terrestrial natural minerals may cause pneumoconiosis and lung cancer, among which iron sulfide is identified as an aggravating factor. In the biological system, iron-sulfur cluster is an inorganic cofactor that is evolutionarily conserved in all the living organisms. Whereas ferrous iron catalyzes the generation of hydroxyl radicals, sulfur is indispensable as a component of antioxidants, such as glutathione. Imbalanced redox homeostasis contributes to oxidative stress, causing ferroptosis, an iron-dependent regulated necrosis characterized by lipid peroxidation, resulting in various disorders. We undertook this study to understand the cellular regulatory mechanisms against major terrestrial minerals containing iron and sulfur from the viewpoint of cellular redox. We used fundamental iron sulfide minerals collected from natural sources to treat human macrophage and fibroblast cells and investigated the biological responses. Alterations in sulfane sulfur, glutathione and iron have been analyzed using either specific fluorescent probes or inductively coupled plasma mass spectrometry. Iron sulfide microparticles with high Fe/S ratio (pyrrhotite; Fe1-XS) induced more reactive sulfane species and glutathione, with less catalytic iron inside cells, whereas the mineral with low Fe/S ratio (pyrite; FeS2) exhibited the opposite effects. Notably both showed cytotoxicity, where pyrite caused ferroptosis but pyrrhotite led to non-ferroptotic disruption. Furthermore, assimilated cellular excess iron was secreted via CD63(+) exosome containing iron-loaded ferritin to the extracellular space with higher iron content in pyrrhotite. Our findings suggest that iron and sulfur work complementarily in maintaining intracellular redox homeostasis, which would be crucial to understand the associated pathology.
Collapse
Affiliation(s)
- Qinying Lyu
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yui Kouketsu
- Department of Earth and Planetary Sciences, Nagoya University Graduate School of Environmental Studies, Furo-cho, Chikusa, Nagoya 484-8601, Japan
| | - Akira Tazaki
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Activity of the Institute of Innovation for Future Society of Nagoya University, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Activity of the Institute of Innovation for Future Society of Nagoya University, Japan
| | - Yashiro Motooka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Center for Low Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Furo-Cho, Chikusa-ku, Nagoya 464-8603, Japan.
| |
Collapse
|
6
|
Liu L, Chen S, Lei Y, Lin Z, Zhou R, Zeng G, Zheng Z, Liu W, Zhou Q, Chen L. REPS2 attenuates cancer stemness through inhibiting Wnt signaling by autophagy mediated degradation of β-catenin. Oncogene 2025:10.1038/s41388-025-03469-y. [PMID: 40514427 DOI: 10.1038/s41388-025-03469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 05/14/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025]
Abstract
Tumor suppressor genes (TSGs) that regulate the stemness of lung cancer cells remain to be determined. We conducted a genome-wide CRISPR/Cas9-mediated screening and identified REPS2 as a potent TSG that negatively regulates the stemness of lung cancer cells. Its tumor suppressive function was confirmed both in vitro and in vivo. Mechanistically, P62 interacts simultaneously with both β-catenin and REPS2, leading to autophagy-lysosome-mediated degradation of β-catenin and attenuation of Wnt signaling. A β-catenin inhibitor synergizes with inhibitors for driver mutants to induce immunogenic cell death, which could be exploited for enhancing efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Lu Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Shuzhen Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Yuxi Lei
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zejian Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Rulan Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Guandi Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zongyao Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Wanting Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Qian Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.
| | - Liang Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Xiao L, Sharma P, Yang X, Abebe D, Loh YP. Neurotrophic Factor-α1/carboxypeptidase E regulates critical protein networks to rescue neurodegeneration, defective synaptogenesis and impaired autophagy in Alzheimer's Disease mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.06.04.657876. [PMID: 40568106 PMCID: PMC12190346 DOI: 10.1101/2025.06.04.657876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
Background The global aging population is increasingly inflicted with Alzheimer's disease (AD), but a cure is still unavailable. Neurotrophic Factor-α1/carboxypeptidase E (NF-α1/CPE) gene therapy has been shown to prevent and reverse memory loss and pathology AD mouse models However, the mechanisms of action of NF-α1/CPE are not fully understood. We investigated if a non-enzymatic form of NF-α1/CPE-E342Q is efficient in reversing AD pathology and carried out a proteomic study to uncover the mechanisms of action of NF-α1/CPE in AD mice. Methods AAV-human NF-α1/CPE and a non-enzymatic form, NF-α1/CPE -E342Q were delivered into hippocampus of 3xTg-AD mice and effects on cognitive function, neurodegeneration, synaptogenesis and autophagy were investigated. A quantitative proteomic analysis of hippocampus of 3xTg-AD mice with and without AAV-NF-α1/CPE treatment was carried out. Results Hippocampal delivery of AAV-NF-α1/CPE-E342Q prevented memory loss, neurodegeneration and increase in activated microglia in 3xTg-AD mice, indicating its action is independent of its enzymatic activity. Quantitative proteomic analysis of hippocampus of 3xTg-AD mice that underwent NF-α1/CPE gene therapy revealed differential expression of >2000 proteins involving many metabolic pathways. Of these, two new proteins down-regulated by NF-α1/CPE: Nexin4 (SNX4) and Trim28 which increase Aβ production and tau levels, respectively were identified. Western blot analysis verified that they were reduced in AAV-NF-α1/CPE treated 3xTg-AD mice compared to untreated mice. Our proteomic analysis indicated synaptic organization as top signaling pathway altered as a response to CPE expression. Synaptic markers PSD95 and Synapsin1 were decreased in 3xTg-AD mice and were restored with AAV-NF-α1/CPE treatment. Proteomic analysis hypothesized involvement of autophagic signaling pathway. Indeed, multiple proteins known to be markers of autophagy were down-regulated in 3xTg-AD mice, accounting for impaired autophagy. Expression of these proteins were upregulated in 3xTg-AD mice with NF-α1/CPE gene therapy, thereby reversing autophagic impairment. Conclusions This study uncovered vast actions of NF-α1/CPE in restoring expression of networks of critical proteins including those necessary for maintaining neuronal survival, synaptogenesis and autophagy, while down-regulating many proteins that promote tau and Aβ accumulation to reverse memory loss and AD pathology in 3xTg-AD mice. AAV-NF-α1/CPE gene therapy uniquely targets many metabolic levels, offering a promising holistic approach for AD treatment. Graphic abstract
Collapse
|
8
|
Al-Hamadani M, Darweesh M, Mohammadi S, Al-Harrasi A. Chloroquine and hydroxychloroquine: Immunomodulatory effects in autoimmune diseases. World J Biol Chem 2025; 16:107042. [DOI: 10.4331/wjbc.v16.i2.107042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/10/2025] [Accepted: 05/07/2025] [Indexed: 05/30/2025] Open
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ), originally developed as antimalarial drugs, have found a new purpose in treating various autoimmune diseases due to their immunomodulatory properties. These drugs work through multiple mechanisms, including inhibiting Toll-like receptor signaling, suppressing antigen presentation, and modulating autophagy. This review article provides a comprehensive analysis of the immunomodulatory effects of CQ and HCQ in several autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, systemic sclerosis, and others. We delve into the intricate mechanisms of action, highlighting the key immune cells involved and discussing the clinical implications of these drugs in managing autoimmune conditions. Our review covers the latest research and clinical trials, offering a comprehensive understanding of the therapeutic potential of CQ and HCQ in autoimmune diseases. We also discuss the challenges and controversies surrounding the use of these drugs, such as their long-term side effects and the need for personalized treatment approaches. By synthesizing current knowledge and identifying areas for future research, this review aims to provide a valuable resource for healthcare professionals and researchers involved in the management of autoimmune diseases.
Collapse
Affiliation(s)
- Moosa Al-Hamadani
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| | - Mahmoud Darweesh
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| | - Saeed Mohammadi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| |
Collapse
|
9
|
Chouhan D, Grossman AS, Kerns KA, Stocke KS, Kim M, Dong PT, Kumar A, Lei L, Lamont RJ, McLean JS, He X, Bor B. Episymbiotic Saccharibacteria suppresses epithelial immunoactivation through Type IV pili and TLR2 dependent endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.30.656655. [PMID: 40501963 PMCID: PMC12157433 DOI: 10.1101/2025.05.30.656655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
Saccharibacteria are episymbionts that require host-bacteria to grow. They are positively associated with inflammatory diseases within the human microbiome, yet their mechanisms for interacting with the human host and contributing to diseases remain unknown. This study investigated interactions between a Saccharibacterium (Nanosynbacter lyticus), its host-bacteria (Schaalia odontolytica), and oral epithelial cells. The host-bacteria induced proinflammatory cytokines in epithelial cells, while Saccharibacteria were immune silent. Remarkably, Saccharibacteria dampened cytokine responses to host-bacteria during coinfection. This effect was driven by Saccharibacteria-induced clustering of TLR2 receptors, a process likely facilitated by type IV, ultimately leading to reduced TLR2-mediated cytokine signalling. High resolution imaging showed that Saccharibacteria were endocytosed by oral epithelial cells, and colocalized with endosome markers, eventually trafficking to lysosomes. Moreover, a subset of the Saccharibacteria survive endocytosis long-term, and retains their capability to reinfect host-bacteria, highlighting a mechanism for persistence in the oral microbiome and a vital role in mammalian immune system modulation.
Collapse
Affiliation(s)
- Deepak Chouhan
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| | - Alex S Grossman
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| | - Kristopher A Kerns
- Department of Periodontics, University of Washington, Seattle WA, 98195, USA
| | - Kendall S Stocke
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Maya Kim
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| | - Pu-Ting Dong
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| | - Ajay Kumar
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| | - Lei Lei
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
- West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, 610093, China
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Jeffrey S McLean
- Department of Periodontics, University of Washington, Seattle WA, 98195, USA
- Department of Oral Health Sciences, University of Washington, Seattle WA, 98195, USA
- Department of Microbiology, University of Washington, Seattle WA, 98109, USA
| | - Xuesong He
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| | - Batbileg Bor
- Department of Microbiology, ADA Forsyth Institute, Somerville MA, 02143, USA
| |
Collapse
|
10
|
Abd El-Fattah AA, Hamid Sadik NA, Shahin AM, Shahin NN. Simvastatin and eugenol restore autophagic flux and alleviate oxidative, inflammatory, and fibrotic perturbations in an arginine-induced chronic pancreatitis rat model. Arch Biochem Biophys 2025; 768:110357. [PMID: 40015469 DOI: 10.1016/j.abb.2025.110357] [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: 10/25/2023] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Chronic pancreatitis (CP), a progressive inflammatory disease characterized by pancreatic tissue destruction and fibrosis, is considered a challenging health burden due to insufficiencies of current management procedures. Autophagy impairment has emerged as a major triggering event in pancreatitis, raising interest in exploring the potential of targeting autophagy as a possible interventional strategy. This study aimed to evaluate the possible ameliorative effect of two autophagy modulators, simvastatin and eugenol, on CP-related perturbations in an arginine-induced rat model. Repeated l-arginine administration (5 g/kg divided into 2 doses with a 1 h interval, given intraperitoneally every 3rd day for a total of 10 times) provoked CP features, demonstrated by acinar damage, oxidative stress, inflammation, and fibrosis. Arginine-triggered pancreatitis was accompanied by hampered pancreatic autophagic flux, evidenced by overexpression of pancreatic p62 and LC3-Ⅱ and downregulation of pancreatic AMPK and LAMP-1 mRNA expression. Treatment with simvastatin (20 mg/kg, intraperitoneally 24 h, before each arginine dose) and eugenol (50 mg/kg/day orally for 30 days) achieved significant anti-oxidative, anti-inflammatory, and anti-fibrotic effects, and reversed the arginine-instigated autophagic blockade, with superior ameliorative effects attained by eugenol. Altogether, simvastatin and eugenol provide a promising interventional approach for CP, at least partly, by restoring the impaired autophagic flux associated with CP.
Collapse
Affiliation(s)
| | | | - Ahmad Mustafa Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nancy Nabil Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
11
|
Pandey A, Goswami A, Jithin B, Shukla S. Autophagy: The convergence point of aging and cancer. Biochem Biophys Rep 2025; 42:101986. [PMID: 40224538 PMCID: PMC11986642 DOI: 10.1016/j.bbrep.2025.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy, a dynamic intracellular degradation system, is critical for cellular renovation and maintaining equilibrium. By eliminating damaged components and recycling essential molecules, autophagy safeguards cellular integrity and function. The versatility of the autophagy process across various biological functions enable cells to adapt and maintain homeostasis under unfavourable conditions. Disruptions in autophagy can shift a cell from a healthy state to a disease state or, conversely, support a return to health. This review delves into the multifaceted role of autophagy during aging and age-related diseases such as cancer, highlighting its significance as a unifying target with promising therapeutic implications. Cancer development is a dynamic process characterized by the acquisition of diverse survival capabilities for proliferating at different stages. This progression unfolds over time, with cancer cells exploiting autophagy to overcome encountered stress conditions during tumor development. Notably, there are several common pathways that utilize the autophagy process during aging and cancer development. This highlights the importance of autophagy as a crucial therapeutic target, holding the potential to not only impede the growth of tumor but also enhance the patient's longevity. This review aims to simplify the intricate relationship between cancer and aging, with a particular focus on the role of autophagy.
Collapse
Affiliation(s)
- Anchala Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | | | | | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
12
|
Ranjana M, Shrilaxmi MS, Nag P, Mitra PK, Sunil D, Dastidar SG, Sudhakar YN, Vennapusa SR, Raju R, Tittonen I, Upadhya D. A versatile electrochemical, colorimetric, and visible light excitable turn-on fluorescent probe for stress-induced H 2S detection. Photochem Photobiol Sci 2025; 24:975-990. [PMID: 40402359 DOI: 10.1007/s43630-025-00739-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Hydrogen sulfide (H2S) holds a distinct role in cell biology. Its level is intricately linked to the homeostasis of the biological environment, underscoring the significance of developing techniques capable of detecting H2S in biological systems. A single probe that offers versatility across different detection techniques opens opportunities for advancements in sensing H2S in various fields. A nitronaphthalimide derivative, NMO prepared using a simple synthetic protocol, has been studied as an electrochemical, colorimetric, and turn-on fluorescence probe for H2S. NMO displayed a detection limit of 9.95 mM and 4.36 mM in the UV-visible and colorimetric studies, respectively, whereas the fluorometric and square wave techniques confirmed lower detection limits of 98.4 μM and 1.24 mM, correspondingly. Further, the real-time imaging of HEK293T cells using NMO during stress-induced autophagy is demonstrated.
Collapse
Affiliation(s)
- M Ranjana
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - M S Shrilaxmi
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Probal Nag
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, 695016, India
| | - Prajoy Kumar Mitra
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, 695016, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Somasish Ghosh Dastidar
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Y N Sudhakar
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sivaranjana Reddy Vennapusa
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, 695016, India
| | - Ramesh Raju
- Department of Electronics and Nanoengineering, Aalto University, P O Box 13500, 00076, Espoo, Finland
| | - Ilkka Tittonen
- Department of Electronics and Nanoengineering, Aalto University, P O Box 13500, 00076, Espoo, Finland
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
13
|
Zhang J, Wang H, Xue X, Wu X, Li W, Lv Z, Su Y, Zhang M, Zhao K, Zhang X, Jia C, Zhu F. Human endogenous retrovirus W family envelope protein (ERVWE1) regulates macroautophagy activation and micromitophagy inhibition via NOXA1 in schizophrenia. Virol Sin 2025:S1995-820X(25)00065-3. [PMID: 40419114 DOI: 10.1016/j.virs.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025] Open
Abstract
The human endogenous retrovirus type W envelope glycoprotein (ERVWE1), located at chromosome 7q21-22, has been implicated in the pathophysiology of schizophrenia. Our previous studies have shown elevated ERVWE1 expression in schizophrenia patients. Growing evidence suggests that autophagy dysfunction contributes to schizophrenia, yet the relationship between ERVWE1 and autophagy remains unclear. In this study, bioinformatics analysis of the human prefrontal cortex RNA microarray dataset (GSE53987) revealed that differentially expressed genes were predominantly enriched in autophagy-related pathways. Clinical data further demonstrated that serum levels of microtubule-associated protein 1 light chain 3β (LC3B), a key marker of macroautophagy, were significantly elevated in schizophrenia patients compared to controls, and positively correlated with ERVWE1 expression. Cellular and molecular experiments suggested that ERVWE1 promoted macroautophagy by increasing the LC3B II/I ratio, enhancing autophagosome formation, and reducing sequestosome 1 (SQSTM1) expression via upregulation of NADPH oxidase activator 1 (NOXA1). Concurrently, NOXA1 downregulated the expression of key micromitophagy-related genes, including PTEN-induced kinase 1 (PINK1), Parkin RBR E3 ubiquitin-protein ligase (Parkin), and the pyruvate dehydrogenase E1 subunit α 1 (PDHA1). As a result, ERVWE1, via NOXA1, inhibited micromitophagy by suppressing the expression of PINK1, Parkin, and PDHA1, thereby leading to impaired production of mitochondrial-derived vesicles (MDVs). Mechanistically, ERVWE1 enhanced NOXA1 transcription by upregulating upstream transcription factor 2 (USF2). In conclusion, ERVWE1 promotes macroautophagy and inhibits micromitophagy through USF2-NOXA1 axis, providing novel mechanistic insight into the role autophagy dysregulation in schizophrenia. These findings suggest that targeting autophagy pathways may offer novel therapeutic strategies for schizophrenia treatment.
Collapse
Affiliation(s)
- Jiahang Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Xing Xue
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiulin Wu
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Wenshi Li
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhao Lv
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yaru Su
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Mengqi Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Kexin Zhao
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xu Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Chen Jia
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
14
|
Dewan S, Sonker H, Chaudhary K, Agrawal S, Chaudhary A, Kumar A, Agrahari B, Singh RG. Self-Assembling Imidazolium Nanoaggregates Trigger a Unique Dynamin-Dependent Cell Death via Cytoplasmic Vacuolization and Mitochondrial Dysfunction in Human Lung Adenocarcinoma. J Med Chem 2025. [PMID: 40408548 DOI: 10.1021/acs.jmedchem.5c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
The identification of alternative cell death pathways is key to developing therapies for apoptosis-resistant cancers. We investigated cell death induced by delocalized lipophilic cation (DLC) nanoaggregates in A549 lung carcinoma cells. These DLCs trigger a dynamin-dependent, nonapoptotic pathway involving cytoplasmic vesicle accumulation and mitochondrial dysfunction. Leveraging the mitochondria-targeting ability of lipophilic cations, we designed and synthesized fluorescent mitochondrion-toxic molecules with potent cytotoxicity against A549, MDA-MB-231, and MCF-7 cells. Dynamic light scattering revealed the nanoaggregate formation of the lead compound, L3, in the RPMI media. L3 inhibited metastasis and clonal expansion, induced vacuole formation post endocytosis, and impaired the mitochondrial function, disrupting ATP levels. This led to mitochondrial permeability transition pore (MPTP) opening and oxidative imbalance via glutathione perturbation. L3 demonstrated strong antitumor activity in vitro and in vivo, showing high potential for treating apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Sayari Dewan
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Himanshu Sonker
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Kajal Chaudhary
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Saloni Agrawal
- Department of Biological Science and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ayushi Chaudhary
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ashwini Kumar
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Bhumika Agrahari
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ritika Gautam Singh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
15
|
Ly T, Pickard B, Pandey A, Yap M, Opara J, Arnold L, Martinez-Rivera N, Rosa-Molinar E, New J, Werner L, Farrokhian N, Gunewardena S, O'Neil M, Bur A, Anant S, Washburn MP, Barnaba C, Ding WX, Thomas SM. TRIM16 mediates secretory autophagy in head and neck cancer-associated fibroblasts. Autophagy 2025:1-24. [PMID: 40383937 DOI: 10.1080/15548627.2025.2508064] [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: 09/25/2024] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025] Open
Abstract
Improving treatment options for head and neck squamous cell carcinoma (HNSCC) requires a deeper understanding of the tumor microenvironment, particularly cancer-associated fibroblasts (CAFs). We previously reported that HNSCC-derived FGF2/bFGF (fibroblast growth factor 2) triggers cytokine release from CAFs via secretory autophagy. Here, using transmission electron microscopy, live-cell imaging, and immunofluorescence, we show that CAF autophagosomes transport cargo, including IL6, to the plasma membrane for secretion. Autophagy in CAFs is constitutive and independent of STAT3, MAPK1/ERK2-MAPK3/ERK1 and phosphoinositide 3-kinase (PI3K) signaling. Despite the significant role of secretory autophagy in CAFs, its molecular machinery has remained elusive. Using both a literature based, and an unbiased approach, we studied the molecular machinery involved in autophagosome trafficking in CAFs. We identified TRIM16, a protein previously reported to traffic to autophagosomes, upregulated in CAFs compared to normal oral fibroblasts. Immunohistochemistry of patient HNSCC stroma revealed co-expression of TRIM16 and LC3B, linking TRIM16 to autophagosome function. An unbiased proteomics profiling of immunoprecipitated LC3B+ vesicles in primary HNSCC CAFs revealed enrichment in trafficking proteins, focal adhesion, and mitochondrial proteins. We demonstrate that SEC22B, SNAP23, VAMP3, and STX4 colocalize with LC3B, IL6, and TRIM16 in CAFs. TRIM16 knockdown reduced autophagosomes at the plasma membrane and decreased IL6 secretion from CAFs. These findings uncover key molecular components involved in autophagy-mediated IL6 secretion in CAFs and suggest potential therapeutic targets for HNSCC.Abbreviations: ACTA2/αSMA: actin alpha 2, smooth muscle; CAF: cancer-associated fibroblasts; CM: conditioned media; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DMSO: dimethylsulfoxide; EGFP: enhanced green fluorescent protein; ELISA: enzyme-linked immunosorbent assay; ER: endoplasmic reticulum; FGF2/bFGF: fibroblast growth factor 2; FGFR: fibroblast growth factor receptor; GO: gene ontology; GORASP2/GRASP55: golgi reassembly stacking protein 2; HMGB1: high mobility group box 1; HNSCC: head and neck squamous cell carcinoma; HPV: human papillomavirus; IL6: interleukin 6; IP: immunoprecipitation; LC-MS/MS: liquid chromatography-mass spectrometry/mass spectrometry; LIR: LC3-interacting region; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; NFs: normal oral fibroblasts; NSCLC: non-small cell lung cancer; PLA: proximity ligation assay; SQSTM1/p62: sequestosome 1; STAT3: signal transducer and activator of transcription 3; SNAP23: synaptosome associated protein 23; SNARE: soluble N-ethyl-maleimide-sensitive factor attachment protein receptor; STX4: syntaxin 4; TEM: transmission electron microscopy; TGFB1: transforming growth factor beta 1; TMA: tissue microarray; TRIM: tri-partite motif; VAMP: vesicle associated membrane protein; VC: vehicle control.
Collapse
Affiliation(s)
- Thuc Ly
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Bailey Pickard
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Avisha Pandey
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Marrion Yap
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Julia Opara
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Levi Arnold
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Noraida Martinez-Rivera
- Microscopy and Analytical Imaging Shared Resource Laboratory, University of Kansas, Lawrence, KS, USA
| | - Eduardo Rosa-Molinar
- Microscopy and Analytical Imaging Shared Resource Laboratory, University of Kansas, Lawrence, KS, USA
- Pharmacology and Toxicology, and Neuroscience Graduate Program, University of Kansas, Lawrence, KS, USA
| | - Jacob New
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lauryn Werner
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nathan Farrokhian
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sumedha Gunewardena
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maura O'Neil
- Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andres Bur
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shrikant Anant
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michael P Washburn
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Carlo Barnaba
- Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Wen-Xing Ding
- Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sufi Mary Thomas
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
16
|
Chen Z, Zhu X, Lu MM, Ou Q, Wang X, Zhao Z, Shen Q, Wang Q, Wang Z, Xu JY, Jin C, Gao F, Wang J, Zhang J, Zhang J, Jin X, Bi Y, Lu L, Xu GT, Tian H. PHOSPHO1 Suppresses Ferroptosis in Retinal Pigment Epithelial Cells by Reducing the Levels of Phosphatidylethanolamine Molecular Species. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2505359. [PMID: 40396905 DOI: 10.1002/advs.202505359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/14/2025] [Indexed: 05/22/2025]
Abstract
Iron-induced lipid peroxidation of phosphatidylethanolamine (PE) species is a key driver of ferroptosis in retinal pigment epithelial (RPE) cells, a process closely associated with age-related macular degeneration (AMD). The previous studies have demonstrated that induced retinal pigment epithelial (iRPE) cells generated by transcription factor-mediated reprogramming exhibit superior therapeutic efficacy in treating AMD. In this study, it is found that these iRPE cells are resistant to ferroptosis and further identified phosphoethanolamine/phosphocholine phosphatase 1 (PHOSPHO1) as a critical regulator underlying ferroptosis resistance. Mechanistically, PHOSPHO1 inhibits ferroptosis through two distinct mechanisms. First, it reduces PE levels in the endoplasmic reticulum, thereby limiting PE-derived lipid peroxidation. Second, it suppresses autophagy and ferritinophagy, leading to a reduction in intracellular free iron accumulation. Experiments using an in vivo rat model confirm that PHOSPHO1 effectively protects RPE cells from ferroptotic damage. These findings highlight PHOSPHO1 as a potential therapeutic target for AMD, providing insights into novel ferroptosis-based intervention strategies.
Collapse
Affiliation(s)
- Zhiyang Chen
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoman Zhu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Michael Mingze Lu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qingjian Ou
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Physiology and Pharmacology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xueying Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhenzhen Zhao
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qi Shen
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qian Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhe Wang
- Department of Physiology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Jing-Ying Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Caixia Jin
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Furong Gao
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Juan Wang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jingfa Zhang
- The International Eye Research Institute of the Chinese University of Hong Kong (Shenzhen), Shenzhen, 518000, China
| | - Jieping Zhang
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Physiology and Pharmacology, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoliang Jin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanlong Bi
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lixia Lu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Haibin Tian
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Physiology and Pharmacology, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
17
|
Han Y, Dai J, Cheng J, He Y, Zhao C, Li R, Zhang Y, Zhang L, Zhou T, Shi Y. Cadmium induces autophagy via IRE1 signaling pathway activated by Ca 2 + in GC-2spd cells. Reprod Toxicol 2025; 135:108950. [PMID: 40398541 DOI: 10.1016/j.reprotox.2025.108950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Cadmium (Cd), an environmental toxicant, accumulates in the human body and damages the male reproductive system. To investigate the molecular mechanisms underlying Cd-induced reproductive toxicity, we used GC-2spd cells and treated them with CdCl2. Additionally, we added 2-APB (an inhibitor of the IP3R) and STF-083010 (an inhibitor of IRE1) to investigate whether they could ameliorate Cd-induced reproductive toxicity. Confocal microscopy and flow cytometry confirmed that CdCl2-treated GC-2spd cells displayed imbalance of calcium homeostasis, with upregulation of the expression of the IP3R, a key pathway for endoplasmic reticulum (ER) Ca2+ release. Furthermore, the ER stress (ERS) effector protein IRE1 expression was also increased, suggesting that Cd activated ERS and the IRE1 pathway by disrupting calcium homeostasis. Previous studies have shown that ERS induces autophagy. We performed the MDC assay to detect autophagosome formation, revealing increased expression of autophagy-related proteins LC3-II/LC3-I and Beclin-1 in response to Cd treatment. In contrast, treatment with 2-APB and STF-083010 inhibited autophagy and mitigated cell death. This inhibitory effect may be due to 2-APB blocking IP3R-mediated Ca2+ release, alleviating imbalance of calcium homeostasis, while STF-083010 inhibits IRE1, restoring ER homeostasis and reducing autophagy. These findings suggest that imbalance of calcium homeostasis activates the IRE1 pathway-mediated ERS, leading to excessive autophagy and male reproductive toxicity. Conversely, the addition of 2-APB and STF-083010 reversed these effects, synergistically restoring intracellular Ca2+ homeostasis and inhibiting ERS to promote cell health. This study provides a new therapeutic strategy for Cd-induced male reproductive disorders.
Collapse
Affiliation(s)
- Yue Han
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Juan Dai
- Wuhan centers for Disease Prevention and Control, China
| | - Jinxin Cheng
- Jiang'an District Center for Disease Prevention and Control in Wuhan, China
| | - Yan He
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Chengkun Zhao
- Ezhou centers for Disease Prevention and Control, China.
| | - Rui Li
- Central China Normal University, China
| | - Yaqin Zhang
- Geriatric Hospital Affiliated with Wuhan University of Science and Technology, China
| | - Ling Zhang
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Ting Zhou
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China
| | - Yuqin Shi
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, China.
| |
Collapse
|
18
|
Vignon AN, Dudon G, Oliva G, Thirard S, Alenda UG, Brugoux A, Cazevieille C, Imbert J, Bellières C, Lehmann S, Crozet C, Torrent J, Bertaso F, Le Merrer J, Becker JAJ, Perrier V. Lifelong exposure to polystyrene-nanoplastics induces an attention-deficit hyperactivity disorder-like phenotype and impairs brain aging in mice. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138640. [PMID: 40403375 DOI: 10.1016/j.jhazmat.2025.138640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/24/2025]
Abstract
The accumulation of plastic waste in the environment, breaking down into micro- and nanoplastics, poses significant threats to ecosystem and human health. Plastic particles have been detected in human blood, urine, and placental tissue, indicating widespread exposure. While their long-term health impacts remain unclear, developing brains, especially in fetuses and children, may be vulnerable, potentially resulting in behavioral changes or neurodevelopmental disorders. This study explores the effects of chronic exposure to 23-nm polystyrene nanoplastics at 10 µg/day/kg in wild-type mice across life stages, using exposure levels reflective of human reality. Maternal exposure disrupted critical developmental milestones in pups. In adulthood, exposed mice exhibited Attention-Deficit Hyperactivity Disorder (ADHD)-like traits, including hyperactivity, increased risk-taking behaviors, and impaired motor learning and executive functions. In aging mice, exposure was associated with a lower epileptic threshold, with developing seizures. These behavioral changes were linked to altered gene and synaptic protein expression associated with ADHD and epilepsy. At the cellular level, lifelong nanoplastic exposure caused lysosomal dysfunctions and increased lipofuscin accumulation, indicative of accelerated brain aging. These findings align with the growing prevalence of ADHD and epilepsy in humans, particularly children and the elderly, emphasizing the urgent need to address plastic pollution and its health implications.
Collapse
Affiliation(s)
- Anaïs N Vignon
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Gaëlle Dudon
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Giulia Oliva
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Steeve Thirard
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Ugo G Alenda
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Agathe Brugoux
- UMR1253, Imaging Brain & Neuropsychiatry iBraiN, Université de Tours, INSERM, CNRS, Tours, France
| | - Chantal Cazevieille
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Jacques Imbert
- MGX-Montpellier GenomiX, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Camille Bellières
- MGX-Montpellier GenomiX, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Sylvain Lehmann
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Carole Crozet
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Joan Torrent
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Federica Bertaso
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France; Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Le Merrer
- UMR1253, Imaging Brain & Neuropsychiatry iBraiN, Université de Tours, INSERM, CNRS, Tours, France
| | - Jérôme A J Becker
- UMR1253, Imaging Brain & Neuropsychiatry iBraiN, Université de Tours, INSERM, CNRS, Tours, France.
| | - Véronique Perrier
- Institut des Neurosciences de Montpellier, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
19
|
Chen Y, Klute S, Sparrer KMJ, Serra-Moreno R. RAB5 is a host dependency factor for the generation of SARS-CoV-2 replication organelles. mBio 2025; 16:e0331424. [PMID: 40167317 PMCID: PMC12077180 DOI: 10.1128/mbio.03314-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a threat due to the emergence of variants with increased transmissibility and enhanced escape from immune responses. Like other coronaviruses before, SARS-CoV-2 likely emerged after its transmission from bats. The successful propagation of SARS-CoV-2 in humans might have been facilitated by usurping evolutionarily conserved cellular factors to execute crucial steps in its life cycle, such as the generation of replication organelles-membrane structures where coronaviruses assemble their replication-transcription complex. In this study, we found that RAB5, which is highly conserved across mammals, is a critical host dependency factor for the replication of the SARS-CoV-2 genome. Our results also suggest that SARS-CoV-2 uses RAB5+ membranes to build replication organelles with the aid of COPB1, a component of the COP-I complex, and that the virus protein NSP6 participates in this process. Hence, targeting NSP6 represents a promising approach to interfere with SARS-CoV-2 RNA synthesis and halt its propagation.IMPORTANCEIn this study, we sought to identify the host dependency factors that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses for the generation of replication organelles: cellular membranous structures that SARS-CoV-2 builds in order to support the replication and transcription of its genome. We uncovered that RAB5 is an important dependency factor for SARS-CoV-2 replication and the generation of replication organelles, and that the viral protein NSP6 participates in this process. Hence, NSP6 represents a promising target to halt SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Yuexuan Chen
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Susanne Klute
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Konstantin Maria Johannes Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Ruth Serra-Moreno
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
20
|
Lee J, Park JR, Lee H, Hong SH, Kim WJ, Eickelberg O, Park SM, Ryu S, Cho SJ, Kim SJ, Yang SR. Fludarabine attenuates inflammation and dysregulated autophagy in alveolar macrophages via inhibition of STAT1/IRF1 pathway. Lab Anim Res 2025; 41:12. [PMID: 40336064 PMCID: PMC12057031 DOI: 10.1186/s42826-025-00245-7] [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: 11/20/2024] [Revised: 04/09/2025] [Accepted: 04/27/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Acute lung injury (ALI), including its most severe form, acute respiratory distress syndrome (ARDS), is a common cause of acute hypoxemic respiratory failure. Although its clinical characteristics have been well characterized, the relevant mechanism remains unclear. An imbalance in autophagy leads to alveolar remodeling and triggers the pathogenesis of ARDS. In this study, we assessed the therapeutic efficacy of the STAT1 inhibitor fludarabine (Fluda) in ALI. C57BL6 mice were exposed to lipopolysaccharide (LPS), and their lung tissues were analyzed via next-generation transcriptome sequencing. RESULTS Western blotting revealed that interferon regulatory factor 1 (IRF1) was highly expressed and STAT1 was phosphorylated following LPS exposure. Fluda significantly decreased the protein expression of STAT1/IRF1 and inhibited the alveolar infiltration of neutrophils and macrophages. Nitric oxide (NO), inducible nitric oxide synthase, tumor necrosis factor-α (TNF-α), interferon-γ, and interleukin-6 (IL-6) release was decreased in the lungs of mice and RAW264.7 macrophages following Fluda treatment. In LPS-induced GFP-LC3 transgenic mice treated with Fluda, the counts of LC3-expressing neutrophils and macrophages in bronchoalveolar (BAL) fluid were significantly decreased. Furthermore, Fluda decreased LC3 and p62 protein expression, thereby inhibiting the release of NO, IL-6, and TNF-α in BAL. In RAW264.7 cells, the inhibition of STAT1/IRF1 by Fluda decreased LPS-induced ERK and NF-κB p65 phosphorylation. CONCLUSIONS The inhibition of STAT1/IRF1 by Fluda plays a pivotal role in modulating dysregulated autophagy by suppressing the MAPK and NF-κB p65 pathways in ALI.
Collapse
Affiliation(s)
- Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Jeong-Ran Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Hanbyeol Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Oliver Eickelberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Semin Ryu
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Sung Joon Cho
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341, Chuncheon, South Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea.
- Institute of Medical Science, School of Medicine, Kangwon National University, Gangwon State, 1 Kanwondaehak-Gil, Chuncheon, 24341, South Korea.
| |
Collapse
|
21
|
Ghanbarpour Houshangi M, Shirakura K, Vestweber D. Tie-2 regulates endothelial morphological responses to shear stress by FOXO1-triggered autophagy. PLoS One 2025; 20:e0322869. [PMID: 40323944 PMCID: PMC12052130 DOI: 10.1371/journal.pone.0322869] [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: 10/04/2024] [Accepted: 03/26/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Endothelial cells respond to flow-induced shear stress by morphological changes, a process which is important for vascular development and physiology. High laminar shear stress activates Tie-2 which supports endothelial junction integrity and protects against vascular leaks and the generation of atherosclerotic plaques. METHODS We have examined the role of Tie-2 and FOXO1 in controlling vascular endothelial cell morphology under physiological shear stress. To address this, we exposed human umbilical vein endothelial cells (HUVECs) transfected with siRNA to 15 dyn/cm2 of shear stress for 24 hours. The resulting cells were analyzed by immunofluorescence staining. RESULTS We found that shear stress-induced activation of Tie-2 is required for endothelial cell alignment and elongation in the direction of flow. Mechanistically, we found that FOXO1 is an essential target downstream of Tie-2, which becomes translocated from the nucleus into the cytosol. There, FOXO1 stimulates the formation of autophagosomes, and both FOXO1 and autophagy stimulation are needed for Tie-2-dependent cell alignment. CONCLUSION In conclusion, laminar fluid shear stress stimulates a novel Tie-2-FOXO1-autophagy signaling axis which is required for endothelial cell alignment. This represents a new mechanism by which Tie-2 contributes to vascular protection under laminar shear stress.
Collapse
Affiliation(s)
| | - Keisuke Shirakura
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
22
|
Ho V, Chung L, Rutland T, Lea V, Lim SH, Abubakar A, Ng W, Lee M, Roberts TL, Chua W, Mackenzie S, Lee CS. Prognostic Value of LC3A Protein Expression Patterns in Rectal Cancer Tumors. Cancers (Basel) 2025; 17:1568. [PMID: 40361494 PMCID: PMC12071802 DOI: 10.3390/cancers17091568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/26/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Autophagy is a conserved self-degradation process by which cells break down and recycle their cellular constituents. This process is activated by various stressors, including nutrient deprivation and DNA damage, and has also been associated with tumor progression. In the present study, we aimed to determine the expression patterns, clinicopathological significance, and prognostic value of the autophagy marker microtubule-associated protein 1 light chain 3 alpha (LC3A)-an essential component of autophagic vacuoles-in rectal cancer. Methods: LC3A reactivity was measured by immunohistochemistry in tumor samples from 243 patients who underwent surgery for rectal cancer. Results: Three distinct patterns of LC3A expression were identified: diffuse cytoplasmic, perinuclear, and stone-like structures (SLS). In Kaplan-Meier survival analyses, patients positive for the SLS pattern of LC3A staining in the tumor periphery (TP) had worse overall survival (OS; p = 0.001) and disease-free survival (DFS; p = 0.030) than those without SLSs in this region, as determined by the log-rank test. When patients were stratified by tumor stage, this result was significant in those with stage T3-T4 (OS, p < 0.001; DFS, p = 0.001) but not T1-T2 disease. Multivariate Cox regression analysis further showed an association between TP-localized LC3A SLS positivity and reduced OS for the overall cohort (hazard ratio [HR] = 2.6313, 95% confidence interval [CI]: 1.090-6.349, p = 0.031) and specifically those in the T3-T4 subgroup (HR = 3.347, 95% CI: 1.657-6.760, p = 0.001). Conclusions: Our findings suggest that positivity for SLSs in the TP may hold clinical value as a biomarker for survival prognosis in rectal cancer patients.
Collapse
Affiliation(s)
- Vincent Ho
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Liping Chung
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Tristan Rutland
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Vivienne Lea
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Stephanie H. Lim
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, NSW 2560, Australia
| | - Askar Abubakar
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Weng Ng
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Mark Lee
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Tara L. Roberts
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
| | - Scott Mackenzie
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (T.R.); (V.L.); (A.A.); (T.L.R.); (W.C.); (S.M.); (C.S.L.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| |
Collapse
|
23
|
Gai S, Meng L, Qin Y, Jiang M. Synthesis and Anticancer Studies of Pt(II) Complex Derived From 4-Phenylthiosemicarbazone. Chem Biodivers 2025; 22:e202402972. [PMID: 39745361 DOI: 10.1002/cbdv.202402972] [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: 11/11/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025]
Abstract
Although cisplatin is widely used as a first-line chemotherapy agent, it has significant side effects. Herein, we synthesized a Pt(II) complex (Pt1) derived from o-vanillin-4-phenylthiosemicarbazone ligand and confirmed its crystal structure by x-ray crystallography. Complex Pt1 exhibited potent anticancer activity against various tested cancer cell lines, with particular efficacy against HepG-2 cells. Further investigations revealed that Pt1 inhibited the growth of HepG-2 cells through multiple mechanisms, including the generation of excessive reactive oxygen species (ROS), induction of DNA damage, enhancement of mitochondrial membrane permeability, promotion of apoptosis, and activation of autophagic cell death.
Collapse
Affiliation(s)
- Shuangshuang Gai
- Institute for History and Culture of Science & Technology, Guangxi Minzu University, Nanning, Guangxi, China
| | - Lili Meng
- School of Biological and Food engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| | - Yiming Qin
- School of Biological and Food engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| | - Ming Jiang
- School of Biological and Food engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi, China
| |
Collapse
|
24
|
Bai H, Xi G, Cheng Y. Prostaglandin G/H synthase 1 promotes thrombosis in atrial fibrillation through modulation of platelet activation, macrophage infiltration, inflammation, and autophagy inhibition. Acta Cardiol 2025; 80:254-265. [PMID: 39963884 DOI: 10.1080/00015385.2025.2467009] [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: 10/29/2024] [Revised: 01/15/2025] [Accepted: 02/09/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Prostaglandin G/H synthase 1 (PTGS1) is known to regulate platelet function and inflammation. However, its role in atrial fibrillation (AF)-related thrombosis is not well understood. This study investigates the role of PTGS1 in AF-associated thrombus formation and its underlying mechanisms. METHODS Left atrial appendage (LAA) tissues were collected from 48 patients undergoing valve replacement surgery, divided into three groups: sinus rhythm (SR), AF with thrombus [AF (+) T (+)], and AF without thrombus [AF (+) T (-)]. PTGS1 expression, platelet activation markers (MPA, sCD40L, and d-dimer), macrophage phenotypes (M1 and M2), inflammatory cytokines (IL-1β, TNF-α, IL-6), and autophagy-related proteins (LC3II and p62) were assessed. Furthermore, the effect of PTGS1 manipulation on autophagy in endocardial endothelial cells (EECs) was examined using cell transfection experiments. RESULTS PTGS1 expression was significantly higher in LAA tissues of AF (+) T (+) patients compared to AF (+) T (-) and SR groups. It was positively correlated with reduced LAA emptying velocity (LAAEV), higher CHA2DS2-VASc scores, and elevated platelet activation markers (MPA, sCD40L, and d-dimer). Data also showed increased M1 macrophage infiltration and higher pro-inflammatory cytokines (IL-1β, TNF-α, IL-6) in AF (+) T (+) patients, with PTGS1 expression strongly linked to these markers. Furthermore, PTGS1 overexpression inhibited autophagy in EECs by decreasing LC3II/LC3I ratio and increasing p62 levels, while PTGS1 knockdown promoted autophagy, protecting against endothelial dysfunction. CONCLUSIONS PTGS1 is overexpressed in AF patients with thrombosis and may play an important role in promoting thrombus formation through enhanced platelet activation, inflammation, and inhibition of autophagy.
Collapse
Affiliation(s)
- Hao Bai
- Department of Ultrasound Diagnosis, Anting Hospital, Shanghai, China
| | - Guiyang Xi
- Department of Ultrasound Diagnosis, Anting Hospital, Shanghai, China
| | - Yangyang Cheng
- Department of Ultrasound Diagnosis, Anting Hospital, Shanghai, China
| |
Collapse
|
25
|
Du H, Mizokami A, Ni J, Zhang S, Yamawaki Y, Sano T, Jimi E, Tanida I, Kanematsu T. Role of Testosterone Signaling in Microglia: A Potential Role for Sex-Related Differences in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413375. [PMID: 40125707 PMCID: PMC12097063 DOI: 10.1002/advs.202413375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is less prevalent in men than in women, although mechanisms remain unclear. Microglia degrade aggregated amyloid β (Aβ) through the lysosomal system, including autophagy. G protein-coupled receptor family C group 6 member A (GPRC6A), predominantly expressed in mouse microglial MG6 cells, is a primary mediator of testosterone signaling. This study examines testosterone's role in modulating Aβ-induced autophagy in microglia. Testosterone promotes Aβ-induced autophagy leading to Aβ clearance in MG6 cells by suppressing extracellular signal-regulated kinase (ERK) phosphorylation and subsequently inhibiting mammalian target of rapamycin (mTOR) activation, which is abrogated by shRNA knockdown of GPRC6A. In in vivo experiments with male 5xFAD AD model mice, Aβ clearance activity is associated with autophagy in microglia and is reduced by orchiectomy, but restored by testosterone supplementation. ERK phosphorylation in the brains of male AD model mice is upregulated by orchiectomy. Therefore, testosterone is involved in autophagy-mediated Aβ clearance in microglia. Aβ accumulation in human brain samples from patients with AD is significantly lower in men than in women, with less pronounced colocalization of Aβ with p62 aggregates, suggesting enhanced autophagic activity in men. In conclusion, testosterone enhances Aβ-induced autophagy in microglia, possibly contributing to lower susceptibility to AD in men.
Collapse
Affiliation(s)
- Haiyan Du
- Department of Cell Biology, Aging Science, and PharmacologyDivision of Oral Biological SciencesFaculty of Dental ScienceKyushu University3‐1‐1 Maidashi, Higashi‐kuFukuoka812‐8582Japan
| | - Akiko Mizokami
- OBT Research CenterFaculty of Dental ScienceKyushu University3‐1‐1 Maidashi, Higashi‐kuFukuoka812‐8582Japan
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and BiotherapyDepartment of BiologySchool of Life ScienceBeijing Institute of TechnologyBeijing100081China
| | - Simeng Zhang
- Key Laboratory of Molecular Medicine and BiotherapyDepartment of BiologySchool of Life ScienceBeijing Institute of TechnologyBeijing100081China
| | - Yosuke Yamawaki
- Department of Advanced PharmacologyDaiichi University of Pharmacy22‐1 Tamagawa‐cho, Minami‐kuFukuoka815‐8511Japan
| | - Tomomi Sano
- Department of Cell Biology, Aging Science, and PharmacologyDivision of Oral Biological SciencesFaculty of Dental ScienceKyushu University3‐1‐1 Maidashi, Higashi‐kuFukuoka812‐8582Japan
| | - Eijiro Jimi
- OBT Research CenterFaculty of Dental ScienceKyushu University3‐1‐1 Maidashi, Higashi‐kuFukuoka812‐8582Japan
- Laboratory of Molecular and Cellular BiochemistryDivision of Oral Biological SciencesKyushu University3‐1‐1 Maidashi, Higashi‐kuFukuoka812‐8582Japan
| | - Isei Tanida
- Department of Cellular and Molecular NeuropathologyJuntendo University Graduate School of MedicineTokyo113‐8421Japan
| | - Takashi Kanematsu
- Department of Cell Biology, Aging Science, and PharmacologyDivision of Oral Biological SciencesFaculty of Dental ScienceKyushu University3‐1‐1 Maidashi, Higashi‐kuFukuoka812‐8582Japan
| |
Collapse
|
26
|
Lu L, Feng Q, Wang S, Ghafar MA, Cheng H, Zhou C, Wang L. miR-278-3p targets ATG16L1 to modulate autophagy and suppresses CLas proliferation in Diaphorina citri. Int J Biol Macromol 2025; 308:142441. [PMID: 40154689 DOI: 10.1016/j.ijbiomac.2025.142441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/13/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The phloem-limited bacterium Candidatus Liberibacter asiaticus (CLas) is the primary cause of citrus Huanglongbing (HLB) and is transmitted by the Asian citrus psyllid, Diaphorina citri. MicroRNAs (miRNAs) are key posttranscriptional regulators involved in various biological processes, yet their role in D. citri's response to CLas infection remains unclear. In this study, we found that autophagy levels were significantly elevated in CLas-infected D. citri compared to non-infected individuals. Modulating autophagy influenced CLas titers, suggesting its role in pathogen suppression. Small RNA sequencing identified differentially expressed miRNAs, with miR-278-3p being significantly downregulated by 72.3%. Functional analyses revealed that miR-278-3p regulates autophagy by targeting ATG16L1. Inhibition of miR-278-3p increased autophagosome formation, whereas its overexpression suppressed autophagy. Dual-luciferase reporter assays confirmed miR-278-3p directly binds to the 3'UTR of ATG16L1, negatively regulating its expression. Notably, miR-278-3p inhibition reduced CLas titers by 48.7%, demonstrating its role in pathogen defense. These findings provide novel insights into the molecular mechanisms underlying insect-pathogen interactions and highlight the potential of miRNA-based strategies for controlling plant diseases transmitted by insect vectors. Understanding these regulatory pathways may lead to innovative pest and disease management approaches in agriculture.
Collapse
Affiliation(s)
- Liuyang Lu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qin Feng
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shimin Wang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Muhammad Adeel Ghafar
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Haokun Cheng
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chenghua Zhou
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Liande Wang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou 350002, China; Key Laboratory of Biopesticides and Chemical Biology, Ministry of Education, Fuzhou 350002, China; College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
27
|
Luis-Ravelo D, Fumagallo-Reading F, Febles-Casquero A, Lopez-Fernandez J, Marcellino DJ, Gonzalez-Hernandez T. Dopamine Receptor D3 Induces Transient, mTORC1-Dependent Autophagy That Becomes Persistent, AMPK-Mediated, and Neuroprotective in Experimental Models of Huntington's Disease. Cells 2025; 14:652. [PMID: 40358175 PMCID: PMC12071662 DOI: 10.3390/cells14090652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Huntington disease's (HD) is a neurodegenerative disorder caused by the expansion of a polyglutamine region (PolyQ) within the huntingtin protein (HTT). Mutated huntingtin (mHTT) is cytotoxic, particularly for striatal medium spiny neurons (MSNs), whose degeneration is the hallmark of HD. Autophagy inducers currently available promote the clearance of toxic proteins. However, due to their low selectivity and the possibility that prolonged autophagy hampers essential processes in unaffected cells, researchers have questioned their benefits in neurodegenerative diseases. Since MSNs express dopamine receptors D2 (DRD2) and D3 (DRD3) and DRD2/DRD3 agonists may activate autophagy, here, we explored how healthy and mHTT-challenged cells respond to prolonged DRD2/DRD3 agonist treatment. Autophagy activation and its effects on mHTT/polyQ clearance were studied in R6/1 mice (a genetic model of HD), their wild-type littermates, and DRD2- and DRD3-HEK cells expressing a pathogenic (Q74) and a non-pathogenic (Q23) polyQ fragment of mHTT treated with the DRD2/DRD3 agonist pramipexole. Two forms of DRD3-mediated autophagy were found: a transient mTORC1-dependent in WT mice and Q23-DRD3-HEK cells and a persistent AMPK-ULK1-activated in R6/1 mice and Q74-DRD3-HEK cells. This also promoted a robust clearance of soluble mHTT/polyQ and neuroprotection in striatal neurons and DRD3-HEK cells. The findings indicate that DRD3-induced autophagy may be a safe, disease-modifying intervention in HD patients.
Collapse
Affiliation(s)
- Diego Luis-Ravelo
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
| | - Felipe Fumagallo-Reading
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
| | - Alejandro Febles-Casquero
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain
| | - Jonathan Lopez-Fernandez
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain
| | - Daniel J. Marcellino
- Department of Medical and Translational Biology, Umeå University, 901 87 Umeå, Sweden;
| | - Tomas Gonzalez-Hernandez
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain
| |
Collapse
|
28
|
Mahvash N, Moradi-Hajidavaloo R, Jafarpour F, Hajian M, Rahimi M, Sanei Ata-abadi N, Sadeghi M, Nasr-Esfahani MH. Induction of autophagy in one-cell stage somatic cell nuclear transfer embryos improves preimplantation embryonic development in goat species. PLoS One 2025; 20:e0314176. [PMID: 40293969 PMCID: PMC12036934 DOI: 10.1371/journal.pone.0314176] [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: 12/20/2023] [Accepted: 11/05/2024] [Indexed: 04/30/2025] Open
Abstract
Autophagy is a lysosome-mediated catabolic pathway that is dependent on the mammalian target of rapamycin (mTOR). It plays a crucial role in the degradation of aged organelles and macromolecules. Several studies have explored the role of autophagy in embryonic genome activation and its significance during the early preimplantation development of mammals. In our study, we showed that autophagy is inhibited in one-cell stage SCNT embryos when compared to fertilized counterparts in goats. Notably, we found that 6-DMAP, a kinase inhibitor, reduces the phosphorylation of ERK1/2.This reduction correlates with a decrease in autophagy levels, as indicated by the presence of LC3 puncta in 6-DMAP treated embryos. To address the inhibition of autophagy in goat SCNT embryos, we induced autophagy using Rapamycin at concentrations of 10 and 100 nM for 6 hours, immediately following chemical activation. This induction led to a significant improvement in the development of goat SCNT embryos, as evidenced by an increased blastocyst rate compared to the control group. Our findings suggest that the induction of autophagy during early hours of one-cell stage embryos is critical for pre-implantation development in goat SCNT embryos warrant further investigation. This research opens new avenues for understanding the role of autophagy in embryonic development and its applications in reproductive biotechnology.
Collapse
Affiliation(s)
- Nasrin Mahvash
- Department of Biology, Faculty of Science and Technology, ACECR Institute of Higher Education (Isfahan), Isfahan, Iran
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Reza Moradi-Hajidavaloo
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Farnoosh Jafarpour
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mehdi Hajian
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Rahimi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Nafiseh Sanei Ata-abadi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Biology, Naghshejahan Higher Education Institute, Isfahan, Iran
| | - Marjan Sadeghi
- Department of Biology, Faculty of Science and Technology, ACECR Institute of Higher Education (Isfahan), Isfahan, Iran
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
29
|
Seo MK, Kim H, Choi AJ, Seog DH, Kho WG, Park SW, Lee JG. Effects of tianeptine on mTORC1-mediated neuronal autophagy in primary rat hippocampal neurons under nutrient deprivation. Sci Rep 2025; 15:14488. [PMID: 40280952 PMCID: PMC12032415 DOI: 10.1038/s41598-025-92988-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/04/2025] [Indexed: 04/29/2025] Open
Abstract
The aim of this study was to investigate the effects of the antidepressant tianeptine on the mechanistic target of rapamycin complex 1(mTORC1)-mediated autophagy pathway in primary hippocampal neurons exposed to B27-deprived conditions. When primary hippocampal neurons were treated with tianeptine at doses of 1, 10, 50, and 100 µM for 3 days under B27-deprived conditions, we observed that it activated autophagy and increased the formation of autophagosomes through the upregulation of autophagic proteins, including autophagy-activating kinase 1 (ULK1), Beclin 1, LC3B-II/I, and p62. And at a concentration of 100 µM tianeptine, the decrease in mTORC1 phosphorylation induced by B27 deprivation was significantly reversed. Changes in the expression of autophagic proteins induced by B27 deprivation were reversed by tianeptine treatment in a concentration-dependent manner, and tianeptine significantly reduced the increase in LC3B membrane number induced by B27 deprivation, an effect that was blocked by pretreatment with rapamycin. In conclusion, tianeptine attenuated the activity of mTORC1-mediated autophagy in primary rat hippocampal neurons under B27-deprived conditions. These results may suggest a novel mechanism by which tianeptine may affect autophagy in neurons.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Hyewon Kim
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea
| | - Ah Jeong Choi
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
| | - Dae-Hyun Seog
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Department of Biochemistry, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Dementia and Neurodegenerative Disease Research Center, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Weon-Gyu Kho
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Parasitology, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea.
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea.
| |
Collapse
|
30
|
Akki AJ, Nanduri S, Patil SV, Das KK, Parvatikar P. Exploring the microRNA-mitochondrial nexus in hepatocellular carcinoma. Mitochondrion 2025; 84:102045. [PMID: 40286975 DOI: 10.1016/j.mito.2025.102045] [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: 10/24/2024] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
MicroRNAs (miRNAs) are double-edged swords in hepatocellular carcinoma (HCC) that play a dual role in disease progression and suppression. The pivotal role of miRNAs in gene regulation emphasizes their potential to disrupt critical cellular processes, including mitochondrial function. Given the indispensable role of mitochondria in energy production, apoptosis, and metabolic control, all of which are central to HCC progression, understanding the miRNA-mitochondria axis is crucial. MiRNAs emerge as pivotal regulators of mitochondrial function, exerting profound influence over HCC progression. This comprehensive review delves into the multifaceted roles of miRNAs in modulating mitochondrial biogenesis, dynamics, and apoptosis. MiRNA impacts key metabolic pathways, including energy metabolism, fatty acid metabolism, and oxidative stress. The intricate interplay between miRNAs and mitochondrial function extends to the regulation of mitophagy and ferroptosis. By exploring the microRNA-mitochondrial axis, this review offers insights for identifying novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Ali Jawad Akki
- Department of Biotechnology, School of Applied Science and Technology, BLDE (Deemed to be University), Vijayapura 586103 Karnataka, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shankargouda V Patil
- Department of Biotechnology, School of Applied Science and Technology, BLDE (Deemed to be University), Vijayapura 586103 Karnataka, India; Department of Pediatrics, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapura 586103 Karnataka, India
| | - Kusal K Das
- Laboratory of Vascular Physiology & Medicine, Department of Physiology, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapura 586103 Karnataka, India
| | - Prachi Parvatikar
- Department of Biotechnology, School of Applied Science and Technology, BLDE (Deemed to be University), Vijayapura 586103 Karnataka, India.
| |
Collapse
|
31
|
Zhong C, Du J, Zhu H, Gao J, Xu G, Xu P. Intestinal Microbiota and Gene Expression Alterations in Chinese Mitten Crab ( Eriocheir sinensis) Under Deltamethrin Exposure. Antioxidants (Basel) 2025; 14:510. [PMID: 40427392 PMCID: PMC12108348 DOI: 10.3390/antiox14050510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 05/29/2025] Open
Abstract
The intestine is an important immune organ of aquatic animals and it plays an essential role in maintaining body health and anti-oxidative stress. To investigate the toxic effects of deltamethrin in intestinal tissue of Chinese mitten crabs (Eriocheir sinensis), 120 healthy crabs were randomly divided into two experimental groups (blank control group and deltamethrin-treated group), with three replicates in each group. After being treated with deltamethrin for 24 h, 48 h, 72 h, and 96 h, intestinal tissues were collected aseptically to assess the effects of deltamethrin on oxidative stress, immunity, apoptosis-related genes, and the structure of microflora in intestinal tissues. Additionally, correlations between gut microbiota composition and intestinal tissue damage-associated genes were analyzed. The results demonstrated that prolonged exposure to deltamethrin induced oxidative stress damage in intestinal tissue. Compared with the blank control group, the expression of autophagy-related genes B-cell lymphoma/Leukemia-2 (bcl-2), c-Jun N-terminal kinase (jnk), Microtuble-associated protein light chain 3 (lc3c), Cysteine-dependent Aspartate-specific Protease 8 (caspase 8), BECN1(beclin1), oxidative stress damage-related genes MAS1 proto-oncogene (mas), Glutathione Peroxidase (gpx), kelch-like ECH-associated protein 1 (keap1), Sequestosome 1 (p62), Interleukin-6 (il-6), and immune-related genes Lipopolysaccharide-induced TNF-alpha Factor (litaf), Heat shock protein 90 (hsp90) and prophenoloxidase (propo) in the deltamethrin treatment group were significantly up-regulated at 96 h (p < 0.05 or p < 0.01). Additionally, 16S rRNA sequencing showed that the diversity of intestinal flora in the deltamethrin-treated group was significantly higher compared with the blank control group (p < 0.01). Analysis of the differences in the composition of intestinal flora at the genus level showed that the relative abundance of Candidatus Bacilloplasma in the deltamethrin treatment group was significantly lower than that in the blank control group (p < 0.01). In contrast, the relative abundances of Flavobacterium, Lachnospiraceae_NK4A136_group, Acinetobacter, Chryseobacterium, Lacihabitans, Taibaiella, Hydrogenophaga, Acidovorax, and Undibacterium were significantly higher than those in the blank control group (p < 0.05 or p < 0.01). Pearson correlation analysis revealed that Malaciobacter, Shewanella, and Prevotella exhibited significant positive correlations with gene indicators (jnk, gpx, lc3c, litaf, hsp90), while Dysgonomonas, Vibrio, and Flavobacterium demonstrated significant negative correlations with multiple gene indicators (caspase 8, p62, il-16, keap1, jnk, etc). These results demonstrate that deltamethrin significantly impacts the gut microbiota, immune function, and antioxidant capacity of E. sinensis. The changes in gut microbiota have correlations with the biomarkers of intestinal tissue injury genes, indicating that gut microbiota plays a crucial role in deltamethrin-induced intestinal tissue damage. These insights contribute to a better understanding of the ecological risks associated with deltamethrin exposure in aquatic organisms.
Collapse
Affiliation(s)
- Chunyi Zhong
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Jinliang Du
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
- International Joint Research Laboratory for Fish Immunopharmacology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Haojun Zhu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Jiancao Gao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Gangchun Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Pao Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| |
Collapse
|
32
|
Rijal R, Gomer RH. Pharmacological inhibition of host pathways enhances macrophage killing of intracellular bacterial pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.06.647500. [PMID: 40291742 PMCID: PMC12026824 DOI: 10.1101/2025.04.06.647500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
After ingestion into macrophage phagosomes, some bacterial pathogens such as Mycobacterium tuberculosis ( Mtb ) evade killing by preventing phagosome acidification and fusion of the phagosome with a lysosome. Mtb accumulates extracellular polyphosphate (polyP), and polyP inhibits macrophage phagosome acidification and bacterial killing. In Dictyostelium discoideum , polyP also inhibits bacterial killing, and we identified some proteins in D. discoideum that polyP requires to suppress the killing of ingested bacteria. Here, we find that pharmacological inhibition of human orthologues of the D. discoideum proteins, including P2Y1 receptors, mammalian Target of Rapamycin (mTOR), and inositol hexakisphosphate kinase, enhances the killing of Mtb , Legionella pneumophila , and Listeria monocytogenes by human macrophages. Mtb inhibits phagosome acidification, expression of the proinflammatory marker CD54, and autophagy, and increases expression of the anti-inflammatory marker CD206. In Mtb -infected macrophages, the polyP-degrading enzyme polyphosphatase (ScPPX) and inhibitors reversed these effects, with ScPPX increasing CD54 expression more in female macrophages compared to male macrophages. In addition, Mtb inhibits proteasome activity, and some, but not all, inhibitors reversed these effects. While the existence of a dedicated polyP signaling pathway remains uncertain, our findings suggest that pharmacological inhibition of select host proteins can restore macrophage function and enhances the killing of intracellular pathogens. Importance Human macrophages engulf bacteria into phagosomes, which then fuse with lysosomes to kill the bacteria. However, after engulfment, pathogenic bacteria such as Mycobacterium tuberculosis , Legionella pneumophila , and Listeria monocytogenes can block phagosome-lysosome fusion, allowing their survival. Here, we show that pharmacological inhibition of specific macrophage proteins reverses these effects and enhances bacterial killing. These findings suggest that targeting host factors involved in these processes may provide a therapeutic strategy to improve macrophage function against infections such as tuberculosis, Legionnaires' disease, and listeriosis.
Collapse
|
33
|
Islam S, Sarkar O, Mukherjee S, Chattopadhyay A. Long-Term Impact of Cr(VI) Exposure in Swiss Albino Mice: ROS-Driven Modulation of Autophagy and Cellular Fate. Biol Trace Elem Res 2025:10.1007/s12011-025-04599-w. [PMID: 40180680 DOI: 10.1007/s12011-025-04599-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Hexavalent chromium [Cr(VI)], due to its high solubility and permeability, is significantly more toxic than trivalent chromium [Cr(III)] as it generates reactive oxygen species (ROS) during cellular reduction. Industrial discharges have led to increasing Cr(VI) contamination in surface and groundwater, posing serious environmental and public health concerns. In our previous study, we demonstrated that exposure to 5 ppm Cr(VI) for 4 and 8 months adversely affected body weight, water consumption, and liver function in Swiss albino mice. Histological analyses revealed tissue alterations, disrupted DNA repair gene expression in liver tissue, and a marked increase in apoptotic gene expression after 8 months of exposure. Building on these findings, we employed the same Cr(VI) concentration (5 ppm via drinking water) over 4 and 8 months in the present study. Our results showed a significant increase in ROS generation in the liver, brain, and kidney tissues at both time intervals. Additionally, the presence of autophagolysosomes was markedly elevated after chronic Cr(VI) exposure in each tissue. We also observed altered expression patterns of key autophagy-related genes (Atg5, Beclin1, and Lc3) and mTor in these tissues. Immunohistochemical analysis further confirmed a significant increase in LC3B expression after 4 months of exposure. Our findings suggest that heightened intracellular oxidative stress triggers a protective autophagy response, mediated via mTOR signaling, to maintain cellular integrity. However, prolonged toxic insult and ROS accumulation may eventually shift pro-survival autophagy toward apoptotic cell death in the liver and brain tissues.
Collapse
Affiliation(s)
- Shehnaz Islam
- Department of Zoology, Visva-Bharati, Santiniketan, 731235, West Bengal, India
| | - Olivia Sarkar
- Department of Zoology, Visva-Bharati, Santiniketan, 731235, West Bengal, India
| | - Sunanda Mukherjee
- Department of Zoology, Visva-Bharati, Santiniketan, 731235, West Bengal, India
| | | |
Collapse
|
34
|
Hosseinzadeh Y, Ghasemzadeh Rahbardar M, Mehri S, Razavi BM, Hosseinzadeh H. Protective effect of aspirin and gentisic acid, a plant-derived phenolic acid, on acrylamide-induced neurotoxicity by inhibiting apoptosis and autophagy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3895-3911. [PMID: 39367985 DOI: 10.1007/s00210-024-03498-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Acrylamide (ACR) is a toxic agent for humans and animals. Gentisic acid, an aspirin metabolite, has antioxidant activity. Therefore, the present study investigated the probable protective effects of aspirin and gentisic acid on ACR-induced neurotoxicity in PC12 cells and rats. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was used to assess the effects of aspirin and gentisic acid (1.25, 2.5, 5 µM) on ACR (5 mM) toxicity. Male Wistar rats were randomly divided into 13 groups: (1) Control group, (2) ACR (50 mg/kg, 11 days, i.p.), (3-5) ACR + aspirin (25, 50, 75 mg/kg, 11 days, p.o.), (6-8) ACR + gentisic acid (25, 50, 75 mg/kg, 11 days, p.o.), (9) ACR + vitamin E (200 mg/kg, every other day, i.p.), (10, 11) Aspirin (75, 100 mg/kg, 11 days, p.o.), (12, 13) Gentisic acid (75, 100 mg/kg, 11 days, p.o.). Behavioral tests were assessed on the final day of the study. In the cerebral cortex, malondialdehyde (MDA), glutathione (GSH), cleaved-caspase-3, and microtubule-associated protein 1A/1B-light chain 3 (LC3) protein levels were evaluated. When compared with the ACR group, aspirin (2.5, 5 µM) and gentisic acid (2.5 µM) significantly enhanced cell viability. In comparison to the control group, ACR induced severe motor impairment, elevated MDA, cleaved-caspase-3, LC3 II/I ratio, and decreased GSH levels in the cerebral cortex of rats. ACR-induced changes were significantly reversed by aspirin and gentisic acid (25 mg/kg). Oxidative stress, apoptosis, and autophagy play important roles in the neurotoxicity of ACR. Aspirin and gentisic acid significantly reduced ACR-induced toxicity by inhibiting the mentioned mechanisms.
Collapse
Affiliation(s)
| | | | - Soghra Mehri
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Targeted Drug Delivery Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Li M, Hu X, Hu X, Gao F, Cui Y, Wei X, Qin Y, An X, Zhao Y, Gao Y. Inhibition of PHB1/PHB2 suppresses atherosclerotic plaque formation by interrupting PI3K/AKT/mTOR signaling. PLoS One 2025; 20:e0320509. [PMID: 40168274 PMCID: PMC11960914 DOI: 10.1371/journal.pone.0320509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are highly conserved proteins belonging to the stomatin-prohibitin flotillin-HflC/K (SPFH) protein superfamily. They are ubiquitously expressed and implicated in the regulation of cell proliferation, migration, and survival. However, the expression and biological functions of PHB1/PHB2 in atherosclerosis (AS) remain unclear. In the present study, an enzyme-linked immunosorbent assay was used to detect PHB1/PHB2 expression in the serum of patients with hyperlipidemia. The potential effect and mechanism of PHB1/PHB2 in apolipoprotein E-deficient (ApoE-/-) mice were also investigated. shRNA-PHB1 and shRNA-PHB2 lentiviruses were engineered and tail vein-injected into ApoE-/- mice fed a high-fat diet. IL-8, a proatherogenic cytokine, was used as an inducer in vitro. The effects of a PHB1/PHB2 knockdown on vascular smooth muscle cell (VSMC) proliferation, migration, and autophagy and endothelial cell (EC) adhesion were evaluated using methyl thiazolyl tetrazolium (MTT), Transwell migration, Boyden chamber, and monocyte adhesion assays, as well as transmission electron microscopy. Compared with the healthy subjects, PHB1/PHB2 expression was elevated in the serum of patients with hyperlipidemia. Animal experiments showed that downregulation of PHBs reduced the area of atherosclerotic lesions, and the expression of cyclinD1, MMP9, and LC3. In addition, in vitro experiments showed that downregulating PHB1/PHB2 expression under inflammatory stimulation reduced the adhesion, proliferation, migration, and autophagy of ECs and VSMCs by inhibiting the PI3K/Akt/mTOR pathway activation. Collectively, our findings showed that PHBs are activly associated with AS progression.
Collapse
Affiliation(s)
- Mei Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaoyan Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xinxin Hu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Fuhua Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Cui
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Yuanhua Qin
- Department of Parasite, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaohua An
- Clinical Laboratory, Dalian Central Hospital, Dalian, China
| | - Ying Zhao
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| | - Ying Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Liaoning Provincial Core Lab of Medical Molecular Biology, Dalian Medical University, Dalian, China
- Molecular Medicine Laboratory, Dalian Medical University, Dalian, China
| |
Collapse
|
36
|
Abd-Elmagid WM, Amr KS, Ahmed HA, Ali D, Abdelhamed A. Autophagy and Premature Graying of Hair: The Role of LC3 as a Biomarker in a Case-Control Study. Dermatol Pract Concept 2025; 15:dpc.1502a4876. [PMID: 40401898 PMCID: PMC12090954 DOI: 10.5826/dpc.1502a4876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 05/23/2025] Open
Abstract
INTRODUCTION Premature graying of hair (PGH) is a common disorder with a multifactorial etiology. Autophagy, which is self-cellular digestion, has been linked to melanin pigment formation; however, the role of autophagy in PGH has not been investigated well. OBJECTIVES The study aimed to evaluate the relationship between PGH and autophagy by measuring gene expression and serum microtubule-associated protein light chain 3 (LC3) concentration. METHODS A case-control study was conducted on 39 PGH patients and 21 controls. Patients clinically diagnosed with PGH and aged <30 years were included in the study. Blood samples were taken to detect LC3B protein by ELISA in the serum of both groups. White hairs from both groups were collected to detect LC3B gene expression by PCR. RESULTS There was a statistically significant difference between the two groups as regards expression levels of the LC3 gene by PCR (P<0.001), with the mean in the control group (0.71± 0.3) lower than in the PGH group (5.1 ± 1.4). Also, there was a positive significant correlation between LC3 concentration and LC3 gene expression in control (r=0.867, P< 0.001) and in PGH patients (r=0.954, P≤0.001). Multivariate logistic regression analysis for PGH predictors using age, sex (female), hemoglobin level, LC3 concentration, and LC3 gene expression revealed that the only predictor of PGH was LC3 gene expression. CONCLUSIONS Premature graying of hair may have a link with autophagy. LC3 gene expression was increased in PGH patients as compared to the control. LC3 gene expression may be an independent predictor of PGH development. Autophagy modulation may be a therapeutic target for PGH.
Collapse
Affiliation(s)
- Wafaa Mohamed Abd-Elmagid
- Dermatology, Venereology and Andrology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Khalda S. Amr
- Medical Molecular Genetics Department, Human Genetics & Genome Research Division (HGGR), National Research Centre (NRC), Egypt
| | - Hoda A. Ahmed
- Medical Molecular Genetics Department, Human Genetics & Genome Research Division (HGGR), National Research Centre (NRC), Egypt
| | - Dina Ali
- Dermatology, Venereology and Andrology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Amr Abdelhamed
- Dermatology, Venereology and Andrology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
37
|
Orefice V, Ceccarelli F, Barbati C, Buoncuore G, Pirone C, Alessandri C, Conti F. Caffeine improves systemic lupus erythematosus endothelial dysfunction by promoting endothelial progenitor cells survival. Rheumatology (Oxford) 2025; 64:1886-1893. [PMID: 39380132 DOI: 10.1093/rheumatology/keae453] [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: 11/19/2023] [Accepted: 06/20/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVE We studied the role of caffeine intake on endothelial function in SLE by assessing its effect on circulating endothelial progenitor cells (EPCs) both ex vivo in SLE patients and in vitro in healthy donors (HD) treated with SLE sera. METHODS We enrolled SLE patients without traditional cardiovascular risks factors. Caffeine intake was evaluated with a 7-day food frequency questionnaire. EPCs percentage was assessed by flow cytometry analysis and, subsequently, EPCs pooled from six HD were co-cultured with caffeine with and without SLE sera. After 7 days, we evaluated cells' morphology and ability to form colonies, the percentage of apoptotic cells by flow cytometry analysis and the levels of autophagy and apoptotic markers by western blot. Finally, we performed a western blot analysis to assess the A2AR/SIRT3/AMPK pathway. RESULTS We enrolled 31 SLE patients, and observed a positive correlation between caffeine intake and circulating EPCs percentage. HD EPCs treated with SLE sera and caffeine showed an improvement in morphology and in number of EPCs colony-forming units in comparison with those incubated without caffeine. Caffeine was able to restore autophagy and apoptotic markers in HD EPCs as before SLE sera treatment. Finally, caffeine treatment was able to significantly reduce A2AR levels, leading to an increase in protein levels of SIRT3 and subsequently AMPK phosphorylation. CONCLUSIONS Caffeine intake positively correlated with the percentage of circulating EPCs in SLE patients; moreover, caffeine in vitro treatment was able to improve EPC survival and vitality through the inhibition of apoptosis and the promotion of autophagy via A2AR/SIRT3/AMPK pathway.
Collapse
Affiliation(s)
- Valeria Orefice
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Cristiana Barbati
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Giorgia Buoncuore
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Carmelo Pirone
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Lupus Clinic, Rheumatology, Department of Clinical, Internal, Anesthesiologic and Cardiovascular, Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
38
|
Zhou P, Zhang Q, Yang Y, Chen D, Jongkaewwattana A, Jin H, Zhou H, Luo R. Avian TRIM13 attenuates antiviral innate immunity by targeting MAVS for autophagic degradation. Autophagy 2025; 21:754-770. [PMID: 39508267 DOI: 10.1080/15548627.2024.2426114] [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: 08/02/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
MAVS (mitochondrial antiviral signaling protein) is a crucial adaptor in antiviral innate immunity that must be tightly regulated to maintain immune homeostasis. In this study, we identified the duck Anas platyrhynchos domesticus TRIM13 (ApdTRIM13) as a novel negative regulator of duck MAVS (ApdMAVS) that mediates the antiviral innate immune response. Upon infection with RNA viruses, ApdTRIM13 expression increased, and it specifically binds to ApdMAVS through its TM domain, facilitating the degradation of ApdMAVS in a manner independent of E3 ligase activity. Furthermore, ApdTRIM13 recruits the autophagic cargo receptor duck SQSTM1 (ApdSQSTM1), which facilitates its interaction with ApdMAVS independent of ubiquitin signaling, and subsequently delivers ApdMAVS to phagophores for degradation. Depletion of ApdSQSTM1 reduces ApdTRIM13-mediated autophagic degradation of ApdMAVS, thereby enhancing the antiviral immune response. Collectively, our findings reveal a novel mechanism by which ApdTRIM13 regulates type I interferon production by targeting ApdMAVS for selective autophagic degradation mediated by ApdSQSTM1, providing insights into the crosstalk between selective autophagy and innate immune responses in avian species.Abbreviation: 3-MA: 3-methyladenine; ATG5: autophagy related 5; baf A1: bafilomycin A1; BECN1: beclin 1; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CARD: caspase recruitment domain; co-IP: co-immunoprecipitation; DEFs: duck embryonic fibroblasts; DTMUV: duck Tembusu virus; eGFP: enhanced green fluorescent protein; hpi: hours post infection; IFIH1/MDA5: interferon induced with helicase C domain 1; IFN: interferon; IKBKE/IKKε: inhibitor of nuclear factor kappa B kinase subunit epsilon; IP: immunoprecipitation; IRF7: interferon regulatory factor 7; ISRE: interferon-stimulated response element; mAb: monoclonal antibody; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; NBR1: NBR1 autophagy cargo receptor; NFKB: nuclear factor kappa B; pAb: polyclonal antibody; poly(I:C): Polyriboinosinic polyribocytidylic acid; RIGI: RNA sensor RIG-I; RLR: RIGI-like-receptor; SeV: sendai virus; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TBK1: TANK binding kinase 1; TCID50: 50% tissue culture infectious dose; TM: tansmembrane; TOLLIP: toll interacting protein; TRIM: tripartite motif containing; UBA: ubiquitin-associated domain; Ub: ubiquitin; VSV: vesicular stomatitis virus; WT: wild type.
Collapse
Affiliation(s)
- Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Qingxiang Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Yueshan Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Dong Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Anan Jongkaewwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Hongbo Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| |
Collapse
|
39
|
Deri E, Kumar Ojha S, Kartawy M, Khaliulin I, Amal H. Multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in autism spectrum disorder. Sci Rep 2025; 15:10878. [PMID: 40158064 PMCID: PMC11954894 DOI: 10.1038/s41598-025-95860-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Our multi-omics study investigated the molecular mechanisms underlying autism spectrum disorder (ASD) using Shank3Δ4-22 and Cntnap2-/- mouse models. Through global- and phospho- proteomics of the mouse cortex, we focused on shared molecular changes and found that autophagy was particularly affected in both models. Global proteomics identified a small number of differentially expressed proteins that significantly impact postsynaptic components and synaptic function, including key pathways such as mTOR signaling. Phosphoproteomics revealed unique phosphorylation sites in autophagy-related proteins such as ULK2, RB1CC1, ATG16L1, and ATG9, suggesting that altered phosphorylation patterns contribute to impaired autophagic flux in ASD. SH-SY5Y cells with SHANK3 gene deletion showed elevated LC3-II and p62 levels, indicating autophagosome accumulation and autophagy initiation, while the reduced level of the lysosomal activity marker LAMP1 suggested impaired autophagosome-lysosome fusion. The study highlights the involvement of reactive nitrogen species and nitric oxide (NO) on autophagy disruption. Importantly, inhibition of neuronal NO synthase (nNOS) by 7-NI normalized autophagy markers levels in the SH-SY5Y cells and primary cultured neurons. We have previously shown that nNOS inhibition improved synaptic and behavioral phenotypes in Shank3Δ4-22 and Cntnap2-/- mouse models. Our multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in ASD but further investigation is needed to prove the full involvement of autophagy in ASD. Our study underscores the need for further examination into the functional consequences of the identified phosphorylation sites, which may offer potential novel therapeutic autophagy-related targets for ASD treatment.
Collapse
Affiliation(s)
- Eden Deri
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Claudio P, Gabriella M. Targeting autophagy in autoimmune glomerular diseases. J Nephrol 2025:10.1007/s40620-025-02267-9. [PMID: 40106213 DOI: 10.1007/s40620-025-02267-9] [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: 08/26/2024] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Autophagy is a natural process whereby damaged or dying parts of a cell are eliminated and recycled. The term autophagy usually refers to macroautophagy, which is one of three types of autophagy, alongside microautophagy and chaperone-mediated autophagy. Autophagy is activated by adenosine monophosphate-activated protein kinase (AMPK) and inhibited by mammalian target of rapamycin (mTOR) through their interference with Unc-51-like kinase 1 (ULK1). Dysregulated autophagy is deeply involved in autoimmune glomerular diseases. Upregulated autophagy can induce inflammation and activate innate and adaptive immunity. However, autophagy may also exert a protective role on podocytes, enhance endothelial cell function, and preserve proximal tubular epithelial cells during ischemic or endotoxic acute kidney injury (AKI). Hydroxychloroquine (HCQ) can downregulate increased autophagy and is widely used in lupus nephritis. HCQ causes alkalinization, which results in vacuolization of lysosomes and inhibition of their functions. By inhibiting autophagic activity, HCQ may reduce inflammation and innate immunity, inhibit the activation of T cells, restore the T helper 17/T regulator balance, restrict the production of pro-inflammatory cytokines, and modulate co-stimulatory molecules. This reduces the risk of flares, spares the dosage of glucocorticoids, improves lupus activity, and prevents the thrombotic effects of anti-phospholipid antibodies. Recent studies showed that HCQ can also reduce proteinuria in IgA nephropathy (IgAN) and membranous nephropathy (MN). Drugs that improve mitochondrial function or enhance autophagy, such as metformin, sodium-glucose co-transporter 2 (SGLT2) inhibitors or mTOR inhibitors, may exert protective effects on podocytes and reduce proteinuria in MN or focal segmental glomerulosclerosis (FSGS).
Collapse
Affiliation(s)
| | - Moroni Gabriella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4 Pieve Emanuele, 20072, Milan, Italy
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Milan, Italy
| |
Collapse
|
41
|
Aligolighasemabadi F, Bakinowska E, Kiełbowski K, Sadeghdoust M, Coombs KM, Mehrbod P, Ghavami S. Autophagy and Respiratory Viruses: Mechanisms, Viral Exploitation, and Therapeutic Insights. Cells 2025; 14:418. [PMID: 40136667 PMCID: PMC11941543 DOI: 10.3390/cells14060418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Respiratory viruses, such as influenza virus, rhinovirus, coronavirus, and respiratory syncytial virus (RSV), continue to impose a heavy global health burden. Despite existing vaccination programs, these infections remain leading causes of morbidity and mortality, especially among vulnerable populations like children, older adults, and immunocompromised individuals. However, the current therapeutic options for respiratory viral infections are often limited to supportive care, underscoring the need for novel treatment strategies. Autophagy, particularly macroautophagy, has emerged as a fundamental cellular process in the host response to respiratory viral infections. This process not only supports cellular homeostasis by degrading damaged organelles and pathogens but also enables xenophagy, which selectively targets viral particles for degradation and enhances cellular defense. However, viruses have evolved mechanisms to manipulate the autophagy pathways, using them to evade immune detection and promote viral replication. This review examines the dual role of autophagy in viral manipulation and host defense, focusing on the complex interplay between respiratory viruses and autophagy-related pathways. By elucidating these mechanisms, we aim to highlight the therapeutic potential of targeting autophagy to enhance antiviral responses, offering promising directions for the development of effective treatments against respiratory viral infections.
Collapse
Affiliation(s)
- Farnaz Aligolighasemabadi
- Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University of Newfoundland, 300 Prince Phillip Dr., St. John’s, NL A1B 3V6, Canada; (F.A.); (M.S.)
| | - Estera Bakinowska
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada; (E.B.); (K.K.)
- Department of Physiology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Kajetan Kiełbowski
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada; (E.B.); (K.K.)
- Department of Physiology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
| | - Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University of Newfoundland, 300 Prince Phillip Dr., St. John’s, NL A1B 3V6, Canada; (F.A.); (M.S.)
| | - Kevin M. Coombs
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Saeid Ghavami
- Division of BioMedical Sciences, Faculty of Medicine, Health Sciences Centre, Memorial University of Newfoundland, 300 Prince Phillip Dr., St. John’s, NL A1B 3V6, Canada; (F.A.); (M.S.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Akademia Śląska, Ul Rolna 43, 40-555 Katowice, Poland
- Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
42
|
Yue Q, Zeng X, Yang M, Chen J, Liu L, Liu H. Bletilla striata polysaccharide induces autophagy through PI3K/AKT signaling pathway to promote the survival of cross-boundary flap in rats. Front Pharmacol 2025; 16:1544932. [PMID: 40129948 PMCID: PMC11931138 DOI: 10.3389/fphar.2025.1544932] [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: 12/13/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction Distal flap necrosis is a common problem in flap transplantation. Bletilla striata polysaccharide (BSP) is the main medicinal component of traditional Chinese medicine Bletilla striata. The purpose of this study was to investigate the mechanism of BSP promoting flap survival. Methods The control group, BSP low, medium and high dose groups, BSP + autophagy inhibitor 3-methyladenine (3-MA) group were designed to establish a model of cross-boundary flap in rat back. After 7 days of postoperative administration, the samples were taken. Results The optimal dose of BSP was determined to be 250 mg/kg/d according to the survival rate of flap, microvessel density, intra-arterial diameter, expression of vascular-related protein and pharmacological toxicity. By detecting the expression level of autophagy-related proteins, it was found that BSP could activate autophagy. After autophagy was blocked, the therapeutic effect of BSP was reversed. In addition, BSP activated the PI3K/AKT signaling pathway. Discussion Studies have shown that BSP induces autophagy by activating PI3K/AKT signaling pathway, thereby promoting angiogenesis and improving survival rate of flap.
Collapse
Affiliation(s)
| | | | | | | | | | - Hui Liu
- Yangtze University Health Science Center, Jingzhou, China
| |
Collapse
|
43
|
Pan TL, Cha JL, Wang H, Zhang JS, Xiao JL, Shen J, Zhou M, Li Y, Ma JZ, Zhao KY, Zhang YK, Xiao P, Gao H. The CRISPR/Cas9-Mediated Knockout of VgrG2 in Wild Pathogenic E. coli to Alleviate the Effects on Cell Damage and Autophagy. Vet Sci 2025; 12:249. [PMID: 40266908 PMCID: PMC11945575 DOI: 10.3390/vetsci12030249] [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: 12/25/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 04/25/2025] Open
Abstract
CRISPR/Cas9, as a well-established gene editing technology, has been applied in numerous model organisms, but its application in wild-type E. coli remains limited. Pathogenic wild-type E. coli, a major cause of foodborne illnesses and intestinal inflammation in humans and animals, poses a significant global public health threat. The valine-glycine repeat protein G (VgrG) is a key virulence factor that enhances E. coli pathogenicity. In this study, PCR was used to identify 50 strains carrying the virulence gene VgrG2 out of 83 wild pathogenic E. coli strains, with only one strain sensitive to kanamycin and spectinomycin. A homologous repair template for VgrG2 was constructed using overlap PCR. A dual-plasmid CRISPR/Cas9 system, combining pTarget (spectinomycin resistance) and pCas (kanamycin resistance) with Red homologous recombination, was then used to induce genomic cleavage and knock out VgrG2. PCR and sequencing confirmed the deletion of a 1708 bp fragment of the VgrG2 gene in wild-type E. coli. IPEC-J2 cells were infected with E. coli-WT and E. coli ∆VgrG2, and treated with the mTOR inhibitor rapamycin to study the effects of VgrG2 on the mTOR signaling pathway. The qPCR results showed that VgrG2 activated the mTOR pathway, suppressed mTOR and p62 mRNA levels, and upregulated the autophagy-related genes and LC3-II protein expression. In conclusion, we utilized CRISPR/Cas9 technology to achieve large-fragment deletions in wild-type E. coli, revealing that VgrG2 activates the mTOR signaling pathway and upregulates autophagy markers. These findings offer new insights into E. coli genome editing and clarifies the pathogenic mechanisms through which VgrG2 induces cellular damage.
Collapse
Affiliation(s)
- Tian-Ling Pan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jin-Long Cha
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Hao Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (H.W.); (M.Z.)
| | - Jing-Song Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jin-Long Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jue Shen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Meng Zhou
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (H.W.); (M.Z.)
| | - Yue Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Jin-Zhi Ma
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Kai-Yuan Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Yong-Kang Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (T.-L.P.); (J.-L.C.); (J.-S.Z.); (J.-L.X.); (J.S.); (Y.L.); (J.-Z.M.); (K.-Y.Z.); (Y.-K.Z.)
| |
Collapse
|
44
|
Ren Z, Cai M, Liu X, Li X, Shi W, Lu H, Shen H, Miao G, Zhou Q, Li H. Omega-3 PUFAs improve cognitive function in heat-stressed mice by enhancing autophagy via inhibition of the phosphorylation of the PI3K-Akt-mTOR pathway. Food Funct 2025; 16:1931-1946. [PMID: 39950918 DOI: 10.1039/d4fo04107k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The adverse effects of elevated temperatures on human health are becoming progressively severe. This research established a mouse model of cognitive dysfunction induced by heat stress to examine the impact of omega-3 PUFAs on the cognitive capabilities of heat-stressed mice. The study also aimed to elucidate the role and potential mechanisms of autophagy regulation in cognitive enhancement through omega-3 PUFAs interventions. Administration of omega-3 PUFAs ameliorated cognitive deficits in heat-stressed mice and increased brain concentrations of these fatty acids. Notably, omega-3 PUFAs significantly protected hippocampal neurons' morphology, quantity, and synaptic architecture in heat-stressed mice. Additionally, omega-3 PUFAs intake reduced the prevalence of damaged mitochondria in the hippocampus and mitigated oxidative harm. Further investigation revealed that heat stress induces autophagy. However, the autophagic process becomes dysfunctional, leading to impaired autophagic activity. Omega-3 PUFAs supplementation markedly augmented hippocampal autophagy in the heat-stressed mice. Moreover, heat stress upregulated the phosphorylation of the PI3K-Akt-mTOR pathway in both the mouse hippocampus and HT22 cells. In contrast, omega-3 PUFAs intake significantly diminished the phosphorylation levels within this pathway, alleviating the autophagic fusion barrier imposed by heat stress and promoting autophagic flux. The findings suggest that omega-3 PUFAs supplementation during heat stress may bolster autophagic function by inhibiting the phosphorylation of the PI3K-Akt-mTOR pathway. This modulation reduces structural and oxidative stress damage, ultimately enhancing cognitive function in mice subjected to heat stress.
Collapse
Affiliation(s)
- Zifu Ren
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
- Medicine-Cardiovascular Dept, PLA No.92493 Hospital, Huludao, China
| | - Mengyu Cai
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Xinyao Liu
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Xin Li
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Wenjing Shi
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Hongtao Lu
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Hui Shen
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Gen Miao
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Qicheng Zhou
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| | - Hongxia Li
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, China.
| |
Collapse
|
45
|
Lai GY, Lee YC, Weng HJ, Lai KH, Hsiang MC, Hsu KY, Liao CP. Discoidin domain receptor inhibitor DDR1-IN-1 induces autophagy and necroptotic cell death in malignant peripheral nerve sheath tumor. Cell Death Discov 2025; 11:83. [PMID: 40025071 PMCID: PMC11873111 DOI: 10.1038/s41420-025-02367-2] [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: 07/30/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a soft tissue sarcoma commonly associated with the tumor-predisposition disorder neurofibromatosis 1. The extracellular matrix collagens contribute to many fibrotic tumors; however, the role of collagen signaling in MPNST was unclear. This study investigated the effects of blocking the interaction between collagens and their receptors in MPNST. We first analyzed the expressions of collagen family proteins in MPNSTs and found an overall increase compared to neurofibroma. Treatment of DDR1-IN-1, a small molecule inhibitor for the collagen receptor discoidin domain receptor, induced a robust MPNST cell death, highlighting the dependence of MPNST survival on collagen signaling. DDR1-IN-1 induced MPNST cell death by activating autophagy and necroptosis signaling. Treatment of necroptosis inhibitors necrostatin-1 or necrosulfonamide reduced the numbers of DDR1-IN-1-induced necrotic cells and autolysosomes, suggesting that the autophagic process depends on necroptosis activation. Combinations of DDR1-IN-1 with other anti-MPNST agents revealed synergistic activities against MPNST. In summary, this study discovered a critical MPNST death signaling induced by the small molecule DDR1-IN-1, which might shed light on future MPNST therapeutic strategies.
Collapse
Affiliation(s)
- Guan-Yi Lai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Cheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hao-Jui Weng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Dermatology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, 23561, Taiwan
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Min-Chen Hsiang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kai-Yu Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Ping Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Cancer Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
| |
Collapse
|
46
|
Wang Y, Su H, Lin X, Dai C, Cheng Q, Deng Z, Yang Y, Pu X. 1,25-(OH) 2D 3 improves SD rats high-altitude pulmonary edema by inhibiting ferroptosis and ferritinophagy in alveolar epithelial cells. J Steroid Biochem Mol Biol 2025; 247:106663. [PMID: 39681240 DOI: 10.1016/j.jsbmb.2024.106663] [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: 08/18/2024] [Revised: 11/21/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND This study investigates the protective effects and potential mechanisms of 1,25-(OH)2D3 against high-altitude pulmonary edema (HAPE). METHODS Hypoxia-induced rats were administered 1,25-(OH)2D3 for 24, 48, and 72 hours, and we observed lung tissue injury and pulmonary edema. Immunohistochemistry (IHC) and Western blot analyses were employed to analyze the expression of markers associated with ferroptosis and ferritinophagy in rat lungs. Metabolomics analysis was conducted to investigate changes in serum lipid metabolites. We validated the mechanism of action of 1,25-(OH)2D3 in type II alveolar epithelial cells induced by hypoxia. RESULTS Our results demonstrated that hypoxic exposure significantly altered sodium-water transport in the lungs, leading to edema formation. The degree of pulmonary edema was most pronounced at 48 hours of hypoxi. Treatment with 1,25-(OH)2D3 improved lung function and reduced the degree of pulmonary edema in hypoxic rats. Hypoxia-induced increases in 4-HNE and MDA levels in the lungs, along with iron accumulation, were observed. Hypoxia also resulted in elevated levels of NCOA4, LC3Ⅱ, and FTH1 proteins in the lungs. Furthermore, treatment with 1,25-(OH)2D3 significantly inhibited ferroptosis and ferritinophagy in the lungs after hypoxia. The levels of lipid metabolites, such as L-Aspartic acid and L-Fucose, were significantly elevated in the serum of hypoxic rats. After 1,25-(OH)2D3 treatment, these levels exhibited a significant reduction. CONCLUSION In hypoxic type II alveolar epithelial cells, 1,25-(OH)2D3 improved hypoxia-induced sodium-water transport, ferroptosis, and ferritinophagy, which were reversed by the autophagy agonist Rapamycin.By modulating ferroptosis and ferritinophagy, 1,25-(OH)2D3 mitigated the deleterious effects of hypoxia on pulmonary function.
Collapse
Affiliation(s)
- Yaxuan Wang
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Hong Su
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Xue Lin
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China.
| | - Chongyang Dai
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Qian Cheng
- Qinghai University, Xining, Qinghai Province 810016, China.
| | | | - Yangyang Yang
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Xiaoyan Pu
- Qinghai University, Xining, Qinghai Province 810016, China.
| |
Collapse
|
47
|
Samadi M, Daryanoosh F, Mojtahedi Z, Samsamy Pour A, Nobari H, Zarifkar AH, Khoramipour K. Resistance Training and Resveratrol Supplementation Improve Cancer Cachexia and Tumor Volume in Muscle Tissue of Male Mice Bearing Colon Cancer CT26 Cell Tumors. Cell Biochem Biophys 2025; 83:619-631. [PMID: 39412707 DOI: 10.1007/s12013-024-01491-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 03/03/2025]
Abstract
Losing muscle functions due to reducing muscle mass and quality is one of the main features of cancer cachexia that impairs patients' quality of life and decrease their survival. This study aimed to investigate the synergistic effects of resistance training and resveratrol supplementation on cachexia induced by CT26 tumors in male mice. Forty-eight mice were divided into eight groups randomly: healthy sedentary vehicle (HSV), healthy exercise vehicle (HEV), healthy sedentary resveratrol (HSR), healthy exercise resveratrol (HER), CT-26 tumor-bearing sedentary vehicle (TSV), CT-26 tumor-bearing exercise vehicle (TEV), CT-26 tumor-bearing sedentary resveratrol (TSR) and CT-26 tumor-bearing exercise resveratrol (TER). Training groups performed ladder climbing with weights tied to their tails, for six weeks. Resveratrol-treated groups received 50 mg/kg daily by gavage. The results showed muscle weight, and mTORC1 phosphorylation decreased in TSV compared to the HSV group. mTORC1 phosphorylation was increased in TER compared to TSV, TEV, and TSR. In addition, AMPK phosphorylation was more elevated in HER compared to HSV, HEV, and HSR. LC3BII/I ratio was higher in TSV than HSV group. Tumor volume was increased in all groups, with the lowest increase in TER group. In tumor tissue, mTORC1 phosphorylation was decreased in TER than in TSV, TEV, and TSR groups; AMPK phosphorylation and LC3BII/I ratio were increased in TSV than in TEV, TSR, and TER groups. In conclusion, the synergistic effect of resistance training and resveratrol supplementation is the most effective in reducing tumor volume. These advantages were mostly in line with molecular findings.
Collapse
Affiliation(s)
- Mahdi Samadi
- Department of Sports Sciences, Shiraz University, Shiraz, Iran
| | | | - Zahra Mojtahedi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Hadi Nobari
- Department of Exercise Physiology, Faculty of Educational Sciences and Psychology, University of Mohaghegh Ardabili, Ardabil, 5619911367, Iran
| | - Amir Hossein Zarifkar
- Cellular and Molecular Biology Research Center, Larestan University of Medical Sciences, Larestan, Iran.
| | - Kayvan Khoramipour
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, 47012, Spain.
| |
Collapse
|
48
|
Zhang YJ, He H, Sawuer G, Ma XK, Ainiwaer Z, Wu DD, Zhang XX, An DQ. Tianxiangdan suppresses foam cell formation by enhancing lipophagy and reduces the progression of atherosclerosis. In Vitro Cell Dev Biol Anim 2025; 61:298-310. [PMID: 39808371 DOI: 10.1007/s11626-024-01004-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/10/2024] [Indexed: 01/16/2025]
Abstract
The aim of this study is to assess the impact of Tianxiangdan (TXD) on lipophagy in foam cells and its underlying mechanism in treating atherosclerosis, particularly focusing on its efficacy in lowering blood lipids. In vivo, ApoE-/- atherosclerosis mouse models were established for group intervention. Blood lipid levels of the mice were measured, lipid deposition and autophagy levels in atherosclerotic plaques were assessed, and co-localization of lipid droplets and autophagosomes was examined. In vitro, human THP-1 cells were induced into macrophages and then transformed into foam cells using ox-LDL induction. Different intervention groups were established. Total cellular cholesterol (TC), free cholesterol (FC), and autophagy levels were assessed, while the morphology and distribution of lipid droplets and autophagosomes in cells were observed using transmission electron microscopy. Western blot analysis was performed to evaluate the expression levels of PI3K, Akt, mTOR, TFEB, LC3II/I, ULK1, ABCA1, and p62. TXD effectively lowers blood lipid levels in ApoE-/- atherosclerotic mice, enhances lipophagy, and reduces lipid accumulation in foam cells and arterial lipid plaques. It achieves this by suppressing the expression of p85, Akt, and mTOR, while activating downstream autophagy signals such as TFEB, LC3II/I, and ULK1. Additionally, TXD reduces the expression of p62 and enhances the expression of the cholesterol transport molecule ABCA1. Our findings indicate that TXD activates lipophagy via the PI3K/Akt/mTOR pathway, leading to a reduction in lipid deposition within foam cells and plaques, thereby mitigating atherosclerosis.
Collapse
Affiliation(s)
- Ya-Jie Zhang
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Huan He
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, China
| | - Guligena Sawuer
- Department of Cardiology, Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xue-Kuan Ma
- Medical Department, Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Zulihumaer Ainiwaer
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Dan-Dan Wu
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Xia-Xia Zhang
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China
| | - Dong-Qing An
- College of Traditional Chinese Medicine, Xinjiang Uygur Autonomous Region, Xinjiang Medical University, Urumqi, 830063, China.
- Traditional Chinese Medicine Inheritance and Innovation Center, Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
- College of Traditional Chinese Medicine, Xinjiang Medical University, No. 567 of Shangde North Road, Shuimogou District, Urumqi, 830000, China.
| |
Collapse
|
49
|
Meyerink BL, Karia KS, Rechtzigel MJ, Patthi PR, Edwards AC, Howard JM, Aaseng ER, Aftab S, Weimer JM, Pilaz LJ. Mutation in Wdr45 leads to early motor dysfunction and widespread aberrant axon terminals in a beta-propeller protein associated neurodegeneration (BPAN) patient-inspired mouse model. Front Neurosci 2025; 19:1545004. [PMID: 40092065 PMCID: PMC11907653 DOI: 10.3389/fnins.2025.1545004] [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: 12/13/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Beta-propeller Protein Associated Neurodegeneration (BPAN) is a devastating neurodevelopmental and neurodegenerative disease linked to variants in WDR45. Currently, there is no cure or disease altering treatment for this disease. This is, in part, due to a lack of insight into early phenotypes of BPAN progression and WDR45's role in establishing and maintaining neurological function. Here we generated and characterized a mouse model bearing a c52C > T BPAN patient variant in Wdr45. We show this mutation ablates WDR45 protein expression and alters autophagy in the brain. Behavioral analysis of these mice revealed characteristic signs of BPAN including cognitive impairment, hyperactivity, and motor decline. We show these behaviors coincide with widespread glial activation and early development of axonal spheroids in multiple neuron subclasses throughout the brain. Several lines of evidence suggest these spheroids arise from axon terminals. Transcriptomic analysis uncovered multiple disrupted pathways in the cortex including genes associated with synapses, neurites, endosomes, endoplasmic reticulum, and ferroptosis. This is supported by accumulation of the iron regulating transferrin receptor 1 (TFRC) and the endoplasmic reticulum resident calreticulin (CALR) in the cortex as these animals age. CALR forms spheroid structures similar to the axonal spheroids seen in these animals. Taken together, our data suggest that WDR45 is necessary for healthy brain function and maintenance of axon terminals. This model opens the door to therapeutics targeting BPAN and further exploration of the role of WDR45 in neuronal function.
Collapse
Affiliation(s)
- Brandon L. Meyerink
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Basic Biomedical Sciences, Sanford School of Medicine at the University of South Dakota, Vermillion, SD, United States
| | - Krishna S. Karia
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | | | - Prithvi R. Patthi
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Ariana C. Edwards
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Jessica M. Howard
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Elizabeth R. Aaseng
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Shamiq Aftab
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Vermillion, SD, United States
| | - Louis-Jan Pilaz
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
50
|
Hu L, Liu J, Peng J, Li X, Huang Z, Zhang C, Fan S. TREM2 Alleviates Neuroinflammation by Maintaining Cellular Metabolic Homeostasis and Mitophagy Activity During Early Inflammation. Diseases 2025; 13:60. [PMID: 39997067 PMCID: PMC11854088 DOI: 10.3390/diseases13020060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
AIMS Inflammation is a pivotal characteristic of neurodegenerative diseases. The triggering receptor expressed on the myeloid cells 2 (TREM2) gene has previously been shown to suppress inflammation by directly inhibiting inflammation-related pathways. Mitochondrial dysfunction has recently emerged as another critical pathological manifestation of neurodegenerative diseases. Although TREM2 is involved in the regulation of cellular energy metabolism and mitochondrial autophagy, its role in the relationship between inflammation and mitochondrial autophagy remains unclear. METHODS In this study, we generated TREM2-overexpressing BV-2 cells and established a neuroinflammatory model with LPS. We compared these cells with wild-type cells in terms of inflammation, metabolism, autophagy, and mitochondria using methods such as RT-qPCR, Western blotting, immunocytochemistry, transmission electron microscopy, and flow cytometry. RESULTS Microglia overexpressing TREM2 exhibited increased resistance to inflammation. Additionally, these cells inhibited the metabolic reprogramming that occurs early in LPS-induced inflammation, reduced ROS release, mitigated mitochondrial damage, maintained a certain level of autophagic activity, and cleared damaged mitochondria. Consequently, they alleviated the inflammation caused by the mitochondrial barrier. CONCLUSIONS ur results suggest that TREM2 can alleviate inflammation by maintaining cellular metabolic homeostasis and mitochondrial autophagy activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shengtao Fan
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming 650108, China; (L.H.); (J.L.); (J.P.); (X.L.); (Z.H.); (C.Z.)
| |
Collapse
|