1
|
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
|
2
|
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
|
3
|
Cai J, Zhou H, Liu M, Zhang D, Lv J, Xue H, Zhou H, Zhang W. Host immunity and intracellular bacteria evasion mechanisms: Enhancing host-directed therapies with drug delivery systems. Int J Antimicrob Agents 2025; 65:107492. [PMID: 40107461 DOI: 10.1016/j.ijantimicag.2025.107492] [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: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Host-directed therapies (HDTs) have been investigated as a potential solution to combat intracellular and drug-resistant bacteria. HDTs stem from extensive research on the intricate interactions between the host and intracellular bacteria, leading to a treatment approach that relies on immunoregulation. To improve the bioavailability and safety of HDTs, researchers have utilized diverse drug delivery systems (DDS) to encapsulate and transport therapeutic agents to target cells. In this review, we first introduce the three mechanisms of bactericidal action and intracellular bacterial evasion: autophagy, reactive oxygen species (ROS), and inflammatory cytokines, with a particular focus on autophagy. Special attention is given to the detailed mechanism of xenophagy in clearing intracellular bacteria, a crucial selective autophagy process that specifically targets and degrades intracellular pathogens. Following this, we present the application of DDS to modulate these regulatory methods for intracellular bacteria elimination. By integrating insights from immunology and nanomedicine, this review highlights the emerging role of DDS in advancing HDTs for intracellular bacterial infections and paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Jiayang Cai
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Han Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Mingwei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Dingjian Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Jingxuan Lv
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Haokun Xue
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Houcheng Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
4
|
Chen W, Liu X, Muñoz VR, Kahn CR. Loss of insulin signaling in microglia impairs cellular uptake of Aβ and neuroinflammatory response exacerbating AD-like neuropathology. Proc Natl Acad Sci U S A 2025; 122:e2501527122. [PMID: 40388612 DOI: 10.1073/pnas.2501527122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
Insulin receptors are present on cells throughout the body, including the brain. Dysregulation of insulin signaling in neurons and astrocytes has been implicated in altered mood, cognition, and the pathogenesis of Alzheimer's disease (AD). To define the role of insulin signaling in microglia, the primary phagocytes in the brain critical for maintenance and damage repair, we created mice with an inducible microglia-specific insulin receptor knockout (MG-IRKO). RiboTag profiling of microglial mRNAs revealed that loss of insulin signaling results in alterations of gene expression in pathways related to innate immunity and cellular metabolism. In vitro, loss of insulin signaling in microglia results in metabolic reprogramming with an increase in glycolysis and impaired uptake of Aβ. In vivo, MG-IRKO mice exhibit alterations in mood and social behavior, and when crossed with the 5xFAD mouse model of AD, the resultant mice exhibit increased levels of Aβ plaque and elevated neuroinflammation. Thus, insulin signaling in microglia plays a key role in microglial cellular metabolism and the ability of the cells to take up Aβ, such that reduced insulin signaling in microglia alters mood and social behavior and accelerates AD pathogenesis. Together, these data indicate key roles of insulin action in microglia and the potential of targeting insulin signaling in microglia in treatment of AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Xiangyu Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Vitor Rosetto Muñoz
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Laboratory of Molecular Biology of Exercise, University of Campinas, Limeira, São Paulo 13484-350, Brazil
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
5
|
Sattari M, Shahaboddin ME, Akhavan Taheri M, Khalili E, Tabatabaei-Malazy O, Goodarzi G, Samavarchi Tehrani S, Meshkani R, Panahi G. Therapeutic potential of fisetin in hepatic steatosis: Insights into autophagy pathway regulation and endoplasmic reticulum stress alleviation in high-fat diet-fed mice. PLoS One 2025; 20:e0322335. [PMID: 40402993 DOI: 10.1371/journal.pone.0322335] [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: 07/27/2024] [Accepted: 03/19/2025] [Indexed: 05/24/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common condition with limited FDA-approved treatments due to its complex pathogenesis. Metabolic stress-induced lipotoxicity triggers the unfolded protein response, leading to the development of NAFLD through inflammation and apoptosis. Moreover, metabolic dysregulation compromises autophagic capacity, impairing effective ERphagy and lipophagy in the liver. Fisetin (FSN), a flavonoid present in various fruits and vegetables, has demonstrated the ability to regulate the processes mentioned above and possesses a range of biological properties. In this study using a high-fat diet-induced NAFLD mouse model, treatment with FSN at a dosage of 80 mg/kg per day for eight weeks resulted in reduced hepatic lipid accumulation. This effect was mediated by modulating ER stress through enhancing autophagic activity, as indicated by decreased expression of GRP78, elf2a, ATF4, and CHOP genes, along with increased AMPK phosphorylation, decreased mTOR expression, and elevated levels of ULK1, ATG5, and Beclin1. Additionally, there was an increase in the LCII/LC3I ratio and a reduction in p62 levels in hepatic tissue. Our findings suggest that FSN exerts its effects by activating the AMPK/mTOR signaling pathway and its downstream targets, underscoring its potential therapeutic advantages in managing NAFLD by targeting autophagy and ER stress pathways.
Collapse
Affiliation(s)
- Mahboobe Sattari
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Esmaeil Shahaboddin
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Akhavan Taheri
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ehsan Khalili
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ozra Tabatabaei-Malazy
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Golnaz Goodarzi
- Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sadra Samavarchi Tehrani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Sharma RK, Sahai R, Singh NC, Maheshwari M, Yadav N, Sarkar J, Mitra K. Ormeloxifene induces mitochondrial fission-mediated pro-death autophagy in colon cancer cells. Biochem Biophys Res Commun 2025; 759:151698. [PMID: 40153998 DOI: 10.1016/j.bbrc.2025.151698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Ormeloxifene (ORM) is a nonsteroidal selective estrogen receptor modulator (SERM), developed by the CSIR-Central Drug Research Institute that is approved as an oral contraceptive. However, it has also shown promising anti-cancer activity, especially in breast cancer. Here, we have investigated the anti-cancer effect of ORM on colon cancer cells and show that its antiproliferative activity is mediated through mitochondrial fission and autophagy-associated cell death. We observed that ORM treatment led to an elevation in autophagy markers like LC3II, Beclin1, and Atg7. Autophagy induction and LC3II turnover were monitored by immunofluorescence staining and confocal microscopy. Transmission electron microscopy results confirmed the formation of autophagosomes and autophagolysosomes. Autophagic flux was confirmed by the increased expression of LC3II in cells co-treated with BafilomycinA1(autophagy inhibitor) and ORM. This was further corroborated using tandem mRFP-GFP-LC3 (tfLC3) transfection in DLD-1 cells. Interestingly, we observed that inhibition of autophagy reduced the apoptotic cell population, suggesting pro-death autophagy. ORM treatment caused notable ultrastructural alterations indicative of cellular stress. Notably, ORM triggered the generation of mitochondrial ROS, associated with increased levels of mitochondrial fission and a decrease in mitochondrial fusion proteins. Changes in mitochondrial dynamics were observed under the TEM, which included reduced mitochondrial size and increased mitochondrial number. Inhibition of mitochondrial fission resulted in enhanced cell survival and a concomitant decrease in the autophagic markers, implying that ORM-induced autophagy depends on mitochondrial fission. Taken together, our findings bring to light a novel mechanism where Ormeloxifene targets mitochondrial dynamics to promote autophagy-associated cell death in colon cancer cells.
Collapse
Affiliation(s)
- Rakesh Kumar Sharma
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility and Research, CSIR - Central Drug Research Institute, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rohit Sahai
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility and Research, CSIR - Central Drug Research Institute, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226 031, India
| | - Nishakumari Chentunarayan Singh
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility and Research, CSIR - Central Drug Research Institute, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226 031, India
| | - Mayank Maheshwari
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India
| | - Nisha Yadav
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jayanta Sarkar
- Division of Cancer Biology, CSIR-Central Drug Research Institute (CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kalyan Mitra
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility and Research, CSIR - Central Drug Research Institute, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226 031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
7
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [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: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
8
|
Xiang X, Huang L, Luo W, Qin L, Bian M, Chen W, Han G, Wang N, Mo G, Zhang C, Zhang Y, Yang H, Lu S, Zhang J, Fu T. Neuromuscular electrical stimulation alleviates stroke-related sarcopenia by promoting satellite cells myogenic differentiation via AMPK-ULK1-Autophagy axis. J Orthop Translat 2025; 52:249-264. [PMID: 40342549 PMCID: PMC12059223 DOI: 10.1016/j.jot.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Background Stroke-related sarcopenia can result in muscle mass loss and muscle fibers abnormality, significantly affecting muscle function. The clinical management of stroke-related sarcopenia still requires further research and investigation. This study aims to explore a promising therapy to restore muscle function and promote muscle regeneration in stroke-related sarcopenia, providing a new theory for stroke-related sarcopenia treatment. Methods Stroke-related sarcopenia rat model was established by using permanent middle cerebral artery occlusion (pMCAO) rat and treated with neuromuscular electrical stimulation (NMES). Electrical stimulation (ES) treatment in vitro was mimicked to test the effect of NMES on muscle regeneration in rat skeletal muscle satellite cells (MuSCs). Catwalk, H&E and Masson's trichrome staining, immunofluorescence, transcriptomic analysis, transmission electron microscopy, MuSCs transfection, autophagy flux detection, quantitative real-time PCR analysis, Co-Immunoprecipitation and Western Blot were used to investigate the role of NMES and its mechanism in stroke-related sarcopenia in vivo. Results After NMES treatment, muscle mass and myogenic differentiation were significantly increased in stroke-related sarcopenia rats. The NMES group had more stable gait, neater footprints, higher muscle wet weight, more voluminous morphology and more regenerated muscle fibers. Additionally, ES treatment induced myogenic differentiation in rat MuSCs in vitro. Transcriptomic analysis also showed that "AMPK signaling pathway" was enriched and genes upregulated in ES-treated cells, revealing ES treatment could activate the autophagy in an AMPK-ULK1-dependent mechanism in MuSCs. Besides, it was also founded that infusion of AMPK or ULK1 inhibitor, knockdown of AMPK or ULK1 in MuSCs could block the effect of myotube formation of ES. Conclusion NMES not only restores muscle function but also enhances myogenic activity and muscle regeneration via AMPK-ULK1 autophagy in stroke-related sarcopenia rats. Our study provides a promising strategy for the treatment of stroke-related sarcopenia. The translational potential of this article This study first demonstrates that NMES alleviates stroke-related sarcopenia by promoting MuSCs differentiation through AMPK-ULK1-autophagy axis. The findings reveal a novel therapeutic mechanism, suggesting that NMES can restore muscle function and enhance regeneration in stroke patients. By combining NMES with MuSCs-based therapies, this approach offers a promising strategy for clinical rehabilitation, potentially improving muscle mass and function in stroke survivors. The translational potential lies in its applicability to non-invasive, cost-effective treatments for sarcopenia, enhancing patients' quality of life.
Collapse
Affiliation(s)
- Xingdong Xiang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lei Huang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lieyang Qin
- Department of Cardiovascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guanjie Han
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ning Wang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guokang Mo
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Cheng Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yongxing Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Jian Zhang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Tengfei Fu
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| |
Collapse
|
9
|
Zhu YF, Zhou XY, Lan C, Wen YP, Fu HJ, Li ZC, Li YP, Li SY, Huang FH, Wang L, Yu L, Qin DL, Wu AG, Wu JM, Zhou XG. Tricin Delays Aging and Enhances Muscle Function via Activating AMPK-Mediated Autophagy in Diverse Model Organisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10246-10264. [PMID: 40243450 DOI: 10.1021/acs.jafc.4c12619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Aging leads to progressive decline in the functions of cells, tissues, and organs, severely affecting muscle performance and overall health, highlighting the urgent need for effective therapeutic agents. This study investigated the antiaging properties of tricin, a flavonoid abundant in grains, using biological models, including human fibroblasts, Caenorhabditis elegans (C. elegans), and mice. Tricin significantly alleviated the senescent phenotype in human fibroblasts induced by D-galactose (D-gal), doxorubicin, and replicative senescence, as evidenced by reduced SA-β-gal activity, downregulated senescence markers (p16, p21), and decreased SASP factors. Mechanistically, tricin binds to AMPK and activates the AMPK-mTOR-p70S6K signaling pathway, promoting autophagy and delaying cellular aging. In vivo, tricin extended lifespan, enhanced stress resistance, and improved mobility in C. elegans through aak-2/AMPK-mediated autophagy. In D-gal-induced aging mice, tricin improved muscle function, reducing p16, p21, and SASP expression in muscle tissues. These findings underscore tricin's potential as a promising antiaging therapeutic via AMPK-mediated autophagy activation.
Collapse
Affiliation(s)
- Yun-Fei Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xing-Yue Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Cai Lan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yong-Ping Wen
- College of Food and Bioengineering, Chengdu University, Chengdu, Sichuan 610106, China
| | - Hai-Jun Fu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhi-Chao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ya-Ping Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shi-Ying Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fei-Hong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
| |
Collapse
|
10
|
Prata RBDS, Pinheiro RO. Cell Death Mechanisms in Mycobacterium abscessus Infection: A Double-Edged Sword. Pathogens 2025; 14:391. [PMID: 40333197 PMCID: PMC12030298 DOI: 10.3390/pathogens14040391] [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: 03/07/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025] Open
Abstract
Infections caused by non-tuberculous mycobacteria (NTM), such as Mycobacterium abscessus, elicit diverse cell death mechanisms including apoptosis, necrosis, and pyroptosis, which play key roles in immunopathogenesis. NTM can manipulate these cell death pathways to evade host immune responses, ensuring their intracellular survival and persistence. Apoptosis may aid in antigen presentation and immune activation, while necrosis and pyroptosis trigger excessive inflammation, leading to tissue damage. Autophagy, a crucial cellular defense mechanism, is often induced in response to NTM infection; however, M. abscessus has evolved mechanisms to inhibit autophagic processes, enhancing its ability to survive within host cells. This manipulation of cell death pathways, particularly the dysregulation of autophagy and ferroptosis, contributes to chronic infection, immune evasion, and tissue damage, complicating disease management. Understanding these mechanisms offers potential therapeutic targets for improving treatment strategies against M. abscessus infections.
Collapse
Affiliation(s)
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro 21040-360, Brazil;
| |
Collapse
|
11
|
Yu H, Wu B, He J, Yi J, Wu W, Wang H, Yang Q, Sun D, Zheng H. Exploring the epigenetic impacts of atrazine in zebrafish: Unveiling mechanisms of neurotoxicity, reproductive toxicity, and implications for human health. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 371:125941. [PMID: 40023241 DOI: 10.1016/j.envpol.2025.125941] [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: 11/21/2024] [Revised: 02/07/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Atrazine (ATZ), a widely utilized herbicide, is notable for its long environmental half-life and high solubility, raising significant concerns regarding its ecological and health impacts. While debates continue over its role as an endocrine disruptor, increasing attention has been directed toward its potential epigenetic effects. Utilizing the zebrafish model, a vertebrate with considerable genetic similarity to humans, provides valuable insights into how ATZ exposure may translate into human health risks. This review systematically examines the differential DNA methylation induced by ATZ's non-competitive inhibition of DNA methyltransferases, miRNA dysregulation resulting from mutations in miRNA processing enzymes, and the complex epigenetic interactions affecting histone modifications. Additionally, potential epigenetic biomarkers for ATZ exposure are proposed, which could advance targeted treatment strategies and improve health risk assessments. This synthesis of current understanding identifies knowledge gaps and guides future research towards a more comprehensive understanding of ATZ's epigenetic mechanisms.
Collapse
Affiliation(s)
- Haiyang Yu
- National & Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jia Yi
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Wei Wu
- Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Da Sun
- National & Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China; Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Hongliang Zheng
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
12
|
Campisi D, Hawkins N, Bonjour K, Wollert T. The Role of WIPI2, ATG16L1 and ATG12-ATG5 in Selective and Nonselective Autophagy. J Mol Biol 2025:169138. [PMID: 40221132 DOI: 10.1016/j.jmb.2025.169138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/24/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Autophagy is a conserved cellular recycling pathway that delivers damaged or superfluous cytoplasmic material to lysosomes for degradation. In response to cytotoxic stress or starvation, autophagy can also sequester bulk cytoplasm and deliver it to lysosomes to regenerate building blocks. In macroautophagy, a membrane cisterna termed phagophore that encloses autophagic cargo is generated. The formation of the phagophore depends on a conserved machinery of autophagy related proteins. The phosphatidylinositol(3)-phosphate binding protein WIPI2 facilitates the transition from phagophore initiation to phagophore expansion by recruiting the ATG12-ATG5-ATG16L1 complex to phagophores. This complex functions as an E3-ligase to conjugate ubiquitin-like ATG8 proteins to phagophore membranes, which promotes tethering of cargo to phagophore membranes, phagophore expansion, maturation and the fusion of autophagosomes with lysosomes. ATG16L1 also has important functions independently of ATG12-ATG5 in autophagy and beyond. In this review, we will summarize the functions of WIPI2 and ATG16L1 in selective and nonselective autophagy.
Collapse
Affiliation(s)
- Daniele Campisi
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - N'Toia Hawkins
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Kennedy Bonjour
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Thomas Wollert
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France.
| |
Collapse
|
13
|
Alimohammadi M, Abolghasemi H, Cho WC, Reiter RJ, Mafi A, Aghagolzadeh M, Hushmandi K. Interplay between LncRNAs and autophagy-related pathways in leukemia: mechanisms and clinical implications. Med Oncol 2025; 42:154. [PMID: 40202565 DOI: 10.1007/s12032-025-02710-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
Autophagy is a conserved catabolic process that removes protein clumps and defective organelles, thereby promoting cell equilibrium. Growing data suggest that dysregulation of the autophagic pathway is linked to several cancer hallmarks. Long non-coding RNAs (lncRNAs), which are key parts of gene transcription, are increasingly recognized for their significant roles in various biological processes. Recent studies have uncovered a strong connection between the mutational landscape and altered expression of lncRNAs in the tumor formation and development, including leukemia. Research over the past few years has emphasized the role of lncRNAs as important regulators of autophagy-related gene expression. These RNAs can influence key leukemia characteristics, such as apoptosis, proliferation, epithelial-mesenchymal transition (EMT), migration, and angiogenesis, by modulating autophagy-associated signaling pathways. With altered lncRNA expression observed in leukemia cells and tissues, they hold promise as diagnostic biomarkers and therapeutic targets. The current review focuses on the regulatory function of lncRNAs in autophagy and their involvement in leukemia, potentially uncovering valuable therapeutic targets for leukemia treatment.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Abolghasemi
- Department of Pediatrics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahboobeh Aghagolzadeh
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A, Salehi Z. The role of autophagy in cancer: from molecular mechanism to therapeutic window. Front Immunol 2025; 16:1528230. [PMID: 40248706 PMCID: PMC12003146 DOI: 10.3389/fimmu.2025.1528230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Autophagy is a cellular degradation process that plays a crucial role in maintaining metabolic homeostasis under conditions of stress or nutrient deprivation. This process involves sequestering, breaking down, and recycling intracellular components such as proteins, organelles, and cytoplasmic materials. Autophagy also serves as a mechanism for eliminating pathogens and engulfing apoptotic cells. In the absence of stress, baseline autophagy activity is essential for degrading damaged cellular components and recycling nutrients to maintain cellular vitality. The relationship between autophagy and cancer is well-established; however, the biphasic nature of autophagy, acting as either a tumor growth inhibitor or promoter, has raised concerns regarding the regulation of tumorigenesis without inadvertently activating harmful aspects of autophagy. Consequently, elucidating the mechanisms by which autophagy contributes to cancer pathogenesis and the factors determining its pro- or anti-tumor effects is vital for devising effective therapeutic strategies. Furthermore, precision medicine approaches that tailor interventions to individual patients may enhance the efficacy of autophagy-related cancer treatments. To this end, interventions aimed at modulating the fate of tumor cells by controlling or inducing autophagy substrates necessitate meticulous monitoring of these mediators' functions within the tumor microenvironment to make informed decisions regarding their activation or inactivation. This review provides an updated perspective on the roles of autophagy in cancer, and discusses the potential challenges associated with autophagy-related cancer treatment. The article also highlights currently available strategies and identifies questions that require further investigation in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amir Samii
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Afshin Namdar
- Program in Cell Biology, The Hospital for Sick Children Peter Gilgan Centre for Research and Learning, Toronto, ON, United States
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Ren B, Chen Y, Liu J, Zhou Z, He Y, Wan S, Chen Y, Wu X, Du M, Gao H, Liu L, Shen H. DNA methylation of genes that mediate autophagosome formation contributes to iodine-induced autoimmune thyroiditis: A population-based study conducted at regions with different iodine levels in China. Int J Hyg Environ Health 2025; 265:114537. [PMID: 39965511 DOI: 10.1016/j.ijheh.2025.114537] [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: 10/15/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Autoimmune thyroiditis (AIT) results from a combination of genetic predispositions and environmental factors. This study aims to examine the methylation patterns of genes that mediate autophagosome formation (MAF) between AIT patients and controls from areas with different water iodine levels. METHODS A protein-protein interaction (PPI) network was constructed to explore the interactions of autophagy-related genes (ARGs). The MCODE plugin in Cytoscape software identified two functional epigenetic modules. We included 176 AIT cases from regions with varying water iodine concentrations and matched controls at a 1:1 ratio. The MethylTarget™ assay was employed to assess DNA methylation changes in six MAF-related genes and analyzed the association between iodine levels and epigenetic modifications. RESULTS Significant methylation differences were observed in 11 targets with 75 CpG sites on MAF-related genes between AIT patients and controls (P < 0.05). Methylation levels were generally lower in AIT patients. A negative correlation was found between the methylation status of PRKAB1_36 and urinary iodine concentration (UIC) (rs = -0.208, P = 0.006). In iodine-fortification areas (IFA), AIT patients exhibited significantly higher mRNA expression levels of six MAF-related genes in whole blood compared to controls (P < 0.05). CONCLUSION Environmental water iodine concentration influences the DNA methylation status of MAF-related genes. Hypomethylated targets and sites in these genes were identified, and the relative mRNA expression levels of these genes were significantly increased in AIT cases from regions with a history of prolonged iodine deficiency followed by iodine supplementation.
Collapse
Affiliation(s)
- Bingxuan Ren
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo City, Zhejiang Province 315211, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Yun Chen
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Jinjin Liu
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Zheng Zhou
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Yanhong He
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Siyuan Wan
- Department of Preventive Medicine, Qiqihar Medical University, Qiqihar City, Heilongjiang Province 161006, People's Republic of China
| | - Yao Chen
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Xianhao Wu
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo City, Zhejiang Province 315000, People's Republic of China
| | - Mengxue Du
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Haiyan Gao
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; Department of Clinical Laboratory, The Sixth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province 150023, People's Republic of China
| | - Lixiang Liu
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China
| | - Hongmei Shen
- Centre for Endemic Disease Control, Chinese Centre for Disease Control and Prevention, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China; National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University (23618504) , Harbin City, Heilongjiang Province 150081, People's Republic of China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin City, Heilongjiang Province 150081, People's Republic of China.
| |
Collapse
|
16
|
Wu B, Gao A, He B, Chen Y, Kong X, Wen F, Gao H. RNA-seq analysis of mitochondria-related genes regulated by AMPK in the human trophoblast cell line BeWo. Animal Model Exp Med 2025; 8:649-661. [PMID: 39445545 PMCID: PMC12008445 DOI: 10.1002/ame2.12475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/11/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND How AMP activated protein kinase (AMPK) signaling regulates mitochondrial functions and mitophagy in human trophoblast cells remains unclear. This study was designed to investigate potential players mediating the regulation of AMPK on mitochondrial functions and mitophagy by next generation RNA-seq. METHODS We compared ATP production in protein kinase AMP-activated catalytic subunit alpha 1/2 (PRKAA1/2) knockdown (AKD) and control BeWo cells using the Seahorse real-time ATP rate test, then analyzed gene expression profiling by RNA-seq. Differentially expressed genes (DEG) were examined by Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Then protein-protein interactions (PPI) among mitochondria related genes were further analyzed using Metascape and Ingenuity Pathway Analysis (IPA) software. RESULTS Both mitochondrial and glycolytic ATP production in AKD cells were lower than in the control BeWo cells (CT), with a greater reduction of mitochondrial ATP production. A total of 1092 DEGs were identified, with 405 upregulated and 687 downregulated. GO analysis identified 60 genes associated with the term 'mitochondrion' in the cellular component domain. PPI analysis identified three clusters of mitochondria related genes, including aldo-keto reductase family 1 member B10 and B15 (AKR1B10, AKR1B15), alanyl-tRNA synthetase 1 (AARS1), mitochondrial ribosomal protein S6 (MRPS6), mitochondrial calcium uniporter dominant negative subunit beta (MCUB) and dihydrolipoamide branched chain transacylase E2 (DBT). CONCLUSIONS In summary, this study identified multiple mitochondria related genes regulated by AMPK in BeWo cells, and among them, three clusters of genes may potentially contribute to altered mitochondrial functions in response to reduced AMPK signaling.
Collapse
Affiliation(s)
- Bin Wu
- Department of Reproductive MedicineCentral Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
| | - Albert Gao
- Department of Physiology and Biophysics, College of MedicineHoward UniversityWashingtonDistrict of ColumbiaUSA
| | - Bin He
- Reproductive Physiology LaboratoryNational Research Institute for Family PlanningBeijingP.R. China
| | - Yun Chen
- Landmark BioWatertownMassachusettsUSA
| | - Xiangfeng Kong
- Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaHunanP.R. China
| | - Fayuan Wen
- Department of Biology, College of Arts and SciencesHoward UniversityWashingtonDistrict of ColumbiaUSA
| | - Haijun Gao
- Department of Physiology and Biophysics, College of MedicineHoward UniversityWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
17
|
Chen Y, Wang Z, Ma Q, Sun C. The role of autophagy in fibrosis: Mechanisms, progression and therapeutic potential (Review). Int J Mol Med 2025; 55:61. [PMID: 39950330 PMCID: PMC11878481 DOI: 10.3892/ijmm.2025.5502] [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: 11/06/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Various forms of tissue damage can lead to fibrosis, an abnormal reparative reaction. In the industrialized countries, 45% of deaths are attributable to fibrotic disorders. Autophagy is a highly preserved process. Lysosomes break down organelles and cytoplasmic components during autophagy. The cytoplasm is cleared of pathogens and dysfunctional organelles, and its constituent components are recycled. With the growing body of research on autophagy, it is becoming clear that autophagy and its associated mechanisms may have a role in the development of numerous fibrotic disorders. However, a comprehensive understanding of autophagy in fibrosis is still lacking and the progression of fibrotic disease has not yet been thoroughly investigated in relation to autophagy‑associated processes. The present review focused on the latest findings and most comprehensive understanding of macrophage autophagy, endoplasmic reticulum stress‑mediated autophagy and autophagy‑mediated endothelial‑to‑mesenchymal transition in the initiation, progression and treatment of fibrosis. The article also discusses treatment strategies for fibrotic diseases and highlights recent developments in autophagy‑targeted therapies.
Collapse
Affiliation(s)
| | | | - Qinghong Ma
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Chao Sun
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| |
Collapse
|
18
|
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
|
19
|
Sun W, Chao G, Wu Q, Xia Y, Shang M, Wei Q, Zhou J, Liao L. Astragaloside IV improves the survival rates of retinal ganglion cells in traumatic optic neuropathy by regulating autophagy mediated by the AMPK-MTOR-ULK signaling pathway. Mol Vis 2025; 31:99-112. [PMID: 40384763 PMCID: PMC12085218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/26/2025] [Indexed: 05/20/2025] Open
Abstract
Purpose Autophagy is involved in the pathological changes of traumatic optic neuropathy (TON), and the regulation of autophagy mediated by the AMPK-mTOR-ULK pathway is a potential therapeutic approach. Astragaloside IV (AS-IV) can regulate autophagy and play a therapeutic role in various diseases. This study aimed to observe the therapeutic effect of astragaloside on TON and the role of AMPK-MTOR-ULK pathway-mediated autophagy in this process. Methods After the TON model was established, varying doses of AS-IV were administered as an intervention. Additionally, compound C (an AMPK inhibitor) or 3-methyladenine (an autophagy inhibitor) was administered intraperitoneally in conjunction with AS-IV. Samples were collected following a 7-day intervention period. Western blot analysis was conducted to measure the protein and phosphorylation levels of AMPK, mTOR, and ULK proteins. Moreover, western blot and quantitative reverse transcription PCR assays were used to quantify LC3 levels in retinal tissue. LC3 immunofluorescence was performed to examine autophagy levels in the ganglion cell layer (GCL), while transmission electron microscopy was employed to observe autophagosomes. Additionally, BRN3A immunofluorescence was used to label retinal ganglion cells (RGCs) in the GCL, and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was used to assess apoptosis within the GCL. Finally, optic nerve conduction function was evaluated using flash visual evoked potentials. Results After 7 days, the phosphorylation levels of AMPK, mTOR, and ULK proteins in retinal tissue exhibited significant changes following TON. AS-IV treatment enhanced LC3 messenger RNA and protein levels in TON model rats, and the autophagy-promoting effect of AS-IV was reversed by 3-methyladenine. Moreover, AS-IV elevated P-AMPK and P-ULK levels while decreasing P-mTOR levels. AS-IV also improved the survival rate of RGCs and reduced the P2 peak latency of flash visual evoked potentials. These effects were attenuated by the AMPK inhibitor compound C. Additionally, AS-IV increased the levels of AKT1 and P-AKT1 while decreasing P-S6RP levels in the retinal tissue of TON model rats. Conclusions AS-IV can increase the survival rate of RGCs and improve visual function after TON, which may be related to the improvement of autophagy by regulating the AMPK-MTORC1-ULK pathway.
Collapse
Affiliation(s)
- Wu Sun
- Department of Ophthalmology, Xiyuan Hospital China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Guojun Chao
- Eye Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Qiong Wu
- Beijing Tongren Hospital, Beijing, China
| | - Yanting Xia
- Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mengqiu Shang
- Eye Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
- Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qiping Wei
- Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jian Zhou
- Beijing University of Chinese Medicine, Beijing, China
- Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liang Liao
- Beijing University of Chinese Medicine, Beijing, China
- Department of Ophthalmology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
20
|
Yao YX, Yao CH, Zhang CY, Peng XZ, Dai S, Yu YJ, Li YZ, Zhang SL, Li YX. Chlorogenic Acid Ameliorates Acetaminophen-Induced Liver Injury Through AMPK/mTOR/ULK1-Mediated Autophagy Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:523-542. [PMID: 40145283 DOI: 10.1142/s0192415x2550020x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Acetaminophen (APAP)-induced liver injury (AILI) is a universal liver disease and the predominant cause of acute liver failure in clinical practice. Autophagy is a highly conserved intracellular degradation pathway, with accumulating evidence indicating its involvement in APAP hepatotoxicity. Notably, the serine/threonine AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/unc-51-like kinase 1 (ULK1) pathway serves as the most classical autophagy pathway and engages in autophagy activation. Thus, pharmacological activation of the AMPK/mTOR/ULK1 pathway has emerged as a critical strategy for addressing AILI. Chlorogenic acid (CGA), a main bioactive constituent isolated from Lonicera japonica Thunb., is an autophagy regulator with potential for AILI therapy. However, whether and how CGA modulates autophagy to antagonize AILI has not yet been elucidated. In the present study, we aim to explore the impact of CGA on AILI, as well as the underlying mechanisms in vitro and in vivo. The results demonstrated that CGA could protect the mice and LO2 cells from oxidative stress and liver injury induced by APAP. Regarding mechanisms, CGA activated the AMPK/mTOR/ULK1 pathway, thereby promoting autophagy. This was evidenced by the degradation of p62/SQSTM1 (hereafter referred to as p62), as well as the up-regulation of LC3B, ATG5, and Beclin1. It is worth noting that the aforementioned, CGA-provided beneficial effects were abrogated by pharmacological inhibition of AMPK with Compound C (CC, an AMPK inhibitor). These [Formula: see text] that CGA alleviates oxidative stress and liver injury induced by APAP, which is contingent upon the regulatory effect of CGA on the AMPK/mTOR/ULK1 axis.
Collapse
Affiliation(s)
- Yu-Xin Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Chen-Hao Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Chao-Yang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Xian-Zhi Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Yu-Jie Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Yan-Zhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Sheng-Lin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| | - Yun-Xia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu 611137, China
| |
Collapse
|
21
|
Wang Z, Zhu Y, Yao Y, Zhang W, Wang B, Wang J, Yang Y, Liu L. Natural products targeting regulated cell deaths for adriamycin-induced cardiotoxicity. Cell Death Discov 2025; 11:112. [PMID: 40118839 PMCID: PMC11928682 DOI: 10.1038/s41420-025-02389-w] [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: 10/04/2024] [Revised: 02/01/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Adriamycin (ADR), as an anti-cancer drug in routine clinical application, is utilized to treat various cancers such as ovarian cancer, hematological malignant tumor, and endometrial carcinoma. However, its serious dose-dependent cardiotoxicity extremely limits its clinical application. Currently, there remains a dearth of therapeutic agents to mitigate ADR-induced cardiotoxicity. Extensive research has demonstrated that ADR can simultaneously trigger various regulated cell death (RCD) pathways, such as apoptosis, autophagy, ferroptosis, necroptosis, and pyroptosis. Therefore, drugs targeting these RCD pathways may represent effective strategies for treating ADR-induced cardiotoxicity. Natural products, with their wide availability, low cost, and diverse pharmacological activities, have increasingly gained attention. Various natural products, including polyphenols, flavonoids, terpenoids, and alkaloids, can target the RCD pathways involved in ADR-induced cardiotoxicity. Furthermore, these natural products have exhibited excellent properties in preclinical studies or in vitro experiments. This review summarizes the mechanisms of RCD in ADR-induced cardiotoxicity and systematically reviews the natural products targeting these RCD pathways. Finally, we propose future research directions of natural products in this field.
Collapse
Affiliation(s)
- Zheng Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yu Yao
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Wenyu Zhang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Bo Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jing Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Liwen Liu
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
22
|
Tao D, Xia Y, Liao Q, Yang X, Zhang L, Xie C. Rapamycin mitigates neurotoxicity of fluoride and aluminum by activating autophagy through the AMPK/mTOR/ULK1 pathway in hippocampal neurons and NG108-15 cells. Sci Rep 2025; 15:9801. [PMID: 40119168 PMCID: PMC11928598 DOI: 10.1038/s41598-025-94648-0] [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: 10/11/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Our previous studies have confirmed that fluoride combined with aluminum (FA) can induce hippocampal neuron damage in the second-generation offspring (F2) of rats; however, the underlying mechanisms remain unclear. In this study, we established an F2 rat model and an NG108-15 cell model to investigate the potential modes of action. The autophagy of F2 rat hippocampal neurons and NG108-15 cells was assessed using transmission electron microscopy and immunofluorescence/immunocytochemistry kit, respectively. Hippocampal morphology was evaluated via hematoxylin-eosin (HE) staining. We measured mRNA levels of AMPK, mTOR, ULK1, and LC3 using quantitative reverse transcription PCR, and protein expressions were analyzed by Western blotting. Following treatment with rapamycin (Rap) in FA-exposed F2 rats and NG108-15 cells, a small number of primary lysosomes and autophagosomes appeared within hippocampal cells, with HE staining indicating a near-normal restoration of pyramidal cell morphology. The quantity, intensity, and volume of green fluorescent spots in the cytoplasm of NG108-15 cells increased as observed through fluorescence microscopy. The mRNA expressions of AMPK, ULK1, and LC3 were upregulated while mTOR expressions were downregulated in NG108-15 cells. Correspondingly, protein levels for AMPK, p-AMPK, ULK1, p-ULK1 along with the LC3-II/LC3-I ratio increased whereas those for mTOR, p-mTOR and p62 decreased significantly. Similar trends regarding both mRNA and protein expression were noted within the hippocampus of F2 rats as well. Activation of the AMPK/mTOR/ULK1 signaling pathway by Rap enhances FA-induced autophagy thereby mitigating neuronal damage.
Collapse
Affiliation(s)
- Dan Tao
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, People's Republic of China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, 561113, People's Republic of China
| | - Ya Xia
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, People's Republic of China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, 561113, People's Republic of China
| | - Qilong Liao
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, Research Center of Emerging Contaminants, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou, 510655, People's Republic of China
| | - Xuemei Yang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, People's Republic of China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, 561113, People's Republic of China
| | - Luwen Zhang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, People's Republic of China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, 561113, People's Republic of China
| | - Chun Xie
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, People's Republic of China.
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, 561113, People's Republic of China.
| |
Collapse
|
23
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
24
|
Pabon A, Bhupana JN, Wong CO. Crosstalk between degradation and bioenergetics: how autophagy and endolysosomal processes regulate energy production. Neural Regen Res 2025; 20:671-681. [PMID: 38886933 PMCID: PMC11433889 DOI: 10.4103/nrr.nrr-d-23-02095] [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: 12/27/2023] [Revised: 03/08/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Cells undergo metabolic reprogramming to adapt to changes in nutrient availability, cellular activity, and transitions in cell states. The balance between glycolysis and mitochondrial respiration is crucial for energy production, and metabolic reprogramming stipulates a shift in such balance to optimize both bioenergetic efficiency and anabolic requirements. Failure in switching bioenergetic dependence can lead to maladaptation and pathogenesis. While cellular degradation is known to recycle precursor molecules for anabolism, its potential role in regulating energy production remains less explored. The bioenergetic switch between glycolysis and mitochondrial respiration involves transcription factors and organelle homeostasis, which are both regulated by the cellular degradation pathways. A growing body of studies has demonstrated that both stem cells and differentiated cells exhibit bioenergetic switch upon perturbations of autophagic activity or endolysosomal processes. Here, we highlighted the current understanding of the interplay between degradation processes, specifically autophagy and endolysosomes, transcription factors, endolysosomal signaling, and mitochondrial homeostasis in shaping cellular bioenergetics. This review aims to summarize the relationship between degradation processes and bioenergetics, providing a foundation for future research to unveil deeper mechanistic insights into bioenergetic regulation.
Collapse
Affiliation(s)
- Angelid Pabon
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | | | - Ching-On Wong
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| |
Collapse
|
25
|
Sinclair LV, Youdale T, Spinelli L, Gakovic M, Langlands AJ, Pathak S, Howden AJM, Ganley IG, Cantrell DA. Autophagy repression by antigen and cytokines shapes mitochondrial, migration and effector machinery in CD8 T cells. Nat Immunol 2025; 26:429-443. [PMID: 40016525 PMCID: PMC11876071 DOI: 10.1038/s41590-025-02090-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/15/2025] [Indexed: 03/01/2025]
Abstract
Autophagy shapes CD8 T cell fate; yet the timing, triggers and targets of this process are poorly defined. Herein, we show that naive CD8 T cells have high autophagic flux, and we identify an autophagy checkpoint whereby antigen receptor engagement and inflammatory cytokines acutely repress autophagy by regulating amino acid transporter expression and intracellular amino acid delivery. Activated T cells with high levels of amino acid transporters have low autophagic flux in amino-acid-replete conditions but rapidly reinduce autophagy when amino acids are restricted. A census of proteins degraded and fueled by autophagy shows how autophagy shapes CD8 T cell proteomes. In cytotoxic T cells, dominant autophagy substrates include cytolytic effector molecules, and amino acid and glucose transporters. In naive T cells, mitophagy dominates and selective mitochondrial pruning supports the expression of molecules that coordinate T cell migration and survival. Autophagy thus differentially prunes naive and effector T cell proteomes and is dynamically repressed by antigen receptors and inflammatory cytokines to shape T cell differentiation.
Collapse
Affiliation(s)
- Linda V Sinclair
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Tom Youdale
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Laura Spinelli
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Milica Gakovic
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alistair J Langlands
- National Phenotypic Screening Centre, School of Life Sciences, University of Dundee, Dundee, UK
| | - Shalini Pathak
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Andrew J M Howden
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Ian G Ganley
- MRC PPU, School of Life Sciences, University of Dundee, Dundee, UK
| | - Doreen A Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
26
|
Ma L, Li K, Guo Y, Liu J, Dong J, Li J, Ren Y, Shi L. Selenium triggers AMPK-mTOR pathway to modulate autophagy related to oxidative stress of sheep Leydig cells. Reprod Biol 2025; 25:100973. [PMID: 39580868 DOI: 10.1016/j.repbio.2024.100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/20/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
The objective of this study was to investigate the effect of oxidative stress induced by excessive Se on autophagy of sheep Leydig cells and its underlying mechanism. Leydig cells isolated from the testis of 8-month-old sheep were purified using a discontinuous Percoll density gradient. Cells were divided into four treatment groups (0, 2.0, 4.0 and 8.0 μmol/L of Se). After treatment with Se for 48 h, cell proliferation was detected by CCK-8 assay kit. The biochemical methods were used to evaluate the antioxidant status of Leydig cells. The mRNA transcript and protein abundance related to the AMPK-mTOR pathway and autophagy were detected by real-time PCR and western blot analysis. The results showed that the Leydig cells treated with 8.0 μmol/L Se have the lowest cell viability. The greater ROS content and lower GSH-Px activity were also observed in the Se8.0 group. The inclusion of 2.0 μmol/L Se in the medium did not affect the autophagy of Leydig cells. However, the relative abundance of ATG5 protein and LC3II/I ratio were elevated in the Se8.0 group. Oxidative stress induced by excessive Se (8.0 μmol/L) dramatically improved the abundance of key proteins related to AMPK-mTOR pathway and led to an increase of phosphorylated AMPK, mTOR and ULK1. Compared with the Se8.0 group, compound C could significantly inhibit the key molecules of AMPK-mTOR signaling pathway and mitigate the autophagy of Leydig cells induced by excessive Se. These results indicate that appropriate Se (2.0 μmol/L) can enhance the viability of sheep Leydig cells. Oxidative stress caused by Se excess can induce cell autophagy via activating AMPK-mTOR signaling pathway. The existed crosstalk between autophagy and apoptosis could decide the fate of Leydig cells. This process could play a decisive role in the maintenance of normal male fertility and spermatogenesis by affecting the number of Leydig cells in testis.
Collapse
Affiliation(s)
- Liang Ma
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Kexin Li
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Yaru Guo
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Jinyu Liu
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Jianing Dong
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Jun Li
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Youshe Ren
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, PR China; Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Lei Shi
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, PR China; Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China.
| |
Collapse
|
27
|
Zhang S, Sun J, Yu S, Fu T, Feng Y, Li Z, Zhang D, Wang C. Molecular Observations on the Regulation of hIAPP Aggregation Process and Enhancement of Autophagy by the Short Peptide LPFYPN and Its Modified Peptides of Coix Seed Prolamins. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4659-4672. [PMID: 39950826 DOI: 10.1021/acs.jafc.4c12559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Inhibiting the fibrotic aggregation of the human islet amyloid polypeptide (hIAPP) and accelerating aggregate clearance are crucial factors in type II diabetes regulation. Autophagy plays a central role in hIAPP fibrotic degradation. We investigated how the coix seed prolamin-derived active peptide (LPFYPN, LP6) and its modifying peptides affect hIAPP aggregation and autophagic processes in induced rat insulinoma (INS-1) cells. Both LP6 and its modified peptides inhibited the fibrotic aggregation of hIAPP, an effect related to the binding site within the core region of hIAPP. Additionally, LP6 and the modified peptides reduced hIAPP-induced cytotoxicity, enhanced LC3-II/LC3-I, decreased p62 protein levels, and promoted autophagy by inhibiting the PI3K-Akt-mTOR signaling pathway, thereby upregulating ULK-1 and Beclin-1 expression. Finally, LP6 modified with selenium showed superior inhibition of hIAPP aggregation and cytotoxicity as well as regulation of autophagic flow. These findings emphasize the potential of LP6 and its modified peptides in regulating type II diabetes and other amyloid-related diseases and indicate that they could be further developed as novel functional food ingredients against type 2 diabetes mellitus (T2DM).
Collapse
Affiliation(s)
- Shu Zhang
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
| | - Jingru Sun
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
- National Coarse Cereals Engineering Research Center, Daqing 163319, P. R. China
| | - Shibo Yu
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
- National Coarse Cereals Engineering Research Center, Daqing 163319, P. R. China
| | - Tianxin Fu
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
| | - Yuchao Feng
- Institute of Quality Standards and Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, P. R. China
| | - Zhijiang Li
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
| | - Dongjie Zhang
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
- National Coarse Cereals Engineering Research Center, Daqing 163319, P. R. China
- Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, P. R. China
| | - Changyuan Wang
- College of Food, Heilongjiang Bayi Agricultural University, Xinfeng Lu 5, Daqing 163319, P. R. China
- National Coarse Cereals Engineering Research Center, Daqing 163319, P. R. China
- Heilongjiang Food and Biotechnology Innovation and Research Center (International Cooperation), Daqing 163319, P. R. China
| |
Collapse
|
28
|
Tao M, Zhang LL, Zhou GH, Wang C, Luo X. Inhibition of metabotropic glutamate receptor-5 alleviates hepatic steatosis by enhancing autophagy via activation of the AMPK signaling pathway. World J Gastroenterol 2025; 31:98852. [PMID: 39991675 PMCID: PMC11755260 DOI: 10.3748/wjg.v31.i7.98852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/08/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The global prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has continued to increase annually. Recent studies have indicated that inhibition of metabotropic glutamate receptor 5 (mGluR5) may alleviate hepatic steatosis. However, the precise mechanism warrants further exploration. AIM To investigate the potential mechanism by which mGluR5 attenuates hepatocyte steatosis in vitro and in vivo. METHODS Free fatty acids (FFAs)-stimulated HepG2 cells were treated with the mGluR5 antagonist MPEP and the mGluR5 agonist CHPG. Oil Red O staining and a triglyceride assay kit were used to evaluate lipid content. Western blot analysis was conducted to detect the expression of the autophagy-associated proteins p62 and LC3-II, as well as the expression of the key signaling molecules AMPK and ULK1, in the treated cells. To further elucidate the contributions of autophagy and AMPK, we used chloroquine (CQ) to inhibit autophagy and compound C (CC) to inhibit AMPK activity. In parallel, wild-type mice and mGluR5 knockout (KO) mice fed a normal chow diet or a high-fat diet (HFD) were used to evaluate the effect of mGluR5 inhibition in vivo. RESULTS mGluR5 inhibition by MPEP attenuated hepatocellular steatosis and increased LC3-II and p62 protein expression. The autophagy inhibitor CQ reversed the effects of MPEP. In addition, MPEP promoted AMPK and ULK1 expression in HepG2 cells exposed to FFAs. MPEP treatment led to the nuclear translocation of transcription factor EB, which is known to promote p62 expression. This effect was negated by the AMPK inhibitor CC. mGluR5 KO mice presented reduced body weight, improved glucose tolerance and reduced hyperlipidemia when fed a HFD. Additionally, the livers of HFD-fed mGluR5 KO mice presented increases in LC3-II and p62. CONCLUSION Our results suggest that mGluR5 inhibition promoted autophagy and reduced hepatocyte steatosis through activation of the AMPK signaling pathway. These findings reveal a new functional mechanism of mGluR5 as a target in the treatment of MASLD.
Collapse
Affiliation(s)
- Min Tao
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Li-Li Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Guang-Hong Zhou
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Cong Wang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Xie Luo
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
29
|
Cruz LLD, Sinzato YK, Paula VG, Fioretto MN, Gallego FQ, Barco VS, Camargo ACL, Corrente JE, Justulin LA, Rodrigues T, Volpato GT, Damasceno DC. Maternal hyperglycemia and postnatal high-fat diet impair metabolic regulation and autophagy response in the liver of adult female rats. J Dev Orig Health Dis 2025; 16:e11. [PMID: 39973168 DOI: 10.1017/s204017442400045x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
This study aimed to investigate the mechanisms by which the association between maternal hyperglycemia and postnatal high-fat diet (HFD) exposure compromises metabolic parameters and hepatic autophagy in adult female pups. For this, Sprague Dawley rats, female pups from nondiabetic (control = FC) or diabetic (FD) mothers, were fed a standard diet (SD) or HFD from weaning until adulthood (n minimum = 5 rats/group): FC/SD, FC/HFD, FD/SD, and FD/HFD. In adulthood, these rats were tested with the oral glucose tolerance test, euthanized, and serum biochemistry parameters were analyzed. Liver samples were collected to evaluate cytokines, redox status, and protein expression autophagy and apoptosis markers. Histomorphometric analyses and an assessment of lipofuscin accumulation were also performed to reflect incomplete autolysosomal digestion. The FC/HFD, FD/SD, and FD/HFD groups showed glucose intolerance and an increased number of hepatocytes. Furthermore, FD/SD and FD/HFD rats showed hyperlipidemia and insulin resistance. Adaptations in hepatic redox pathways were observed in the FD/SD group with increased antioxidant defense marker activity. The FD/SD group also exhibited increased autophagy protein expression, such as p-AMPK, LC3-II/LC3-I, and p62/SQSTM1, lipofuscin accumulation, and caspase-3 activation. After exposure to HFD, the adult female pups of diabetic rats had a reduced p-AMPK and LC3-II/LC3-I ratio, the presence of steatosis, oxidative stress, and inflammation. The reduction of autophagy, stimulated by HFD, may be of vital importance for the susceptibility to metabolic dysfunction-associated fatty liver disease induced by maternal diabetes.
Collapse
Affiliation(s)
- Larissa Lopes da Cruz
- Postgraduate Course on Tocogynecology and Laboratory of Experimental Research on Gynecology and Obstetrics - UNIPEX, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Institute of Biological and Health Sciences, Laboratory of System Physiology and Reproductive Toxicology, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Yuri Karen Sinzato
- Postgraduate Course on Tocogynecology and Laboratory of Experimental Research on Gynecology and Obstetrics - UNIPEX, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Verônyca Gonçalves Paula
- Postgraduate Course on Tocogynecology and Laboratory of Experimental Research on Gynecology and Obstetrics - UNIPEX, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Franciane Quintanilha Gallego
- Postgraduate Course on Tocogynecology and Laboratory of Experimental Research on Gynecology and Obstetrics - UNIPEX, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Vinícius Soares Barco
- Postgraduate Course on Tocogynecology and Laboratory of Experimental Research on Gynecology and Obstetrics - UNIPEX, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Ana Carolina Lima Camargo
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Tiago Rodrigues
- Center of Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, São Paulo, Brazil
| | - Gustavo Tadeu Volpato
- Institute of Biological and Health Sciences, Laboratory of System Physiology and Reproductive Toxicology, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Débora Cristina Damasceno
- Postgraduate Course on Tocogynecology and Laboratory of Experimental Research on Gynecology and Obstetrics - UNIPEX, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
30
|
Jalouli M. Emerging Role of Hypoxia-Inducible Factors (HIFs) in Modulating Autophagy: Perspectives on Cancer Therapy. Int J Mol Sci 2025; 26:1752. [PMID: 40004215 PMCID: PMC11855875 DOI: 10.3390/ijms26041752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are master regulators of cellular responses to low oxygen levels and modulate autophagy, a conserved process essential for maintaining homeostasis. Under hypoxic conditions, HIFs regulate the expression of autophagy-related genes and influence autophagic flux and cellular stress responses. Dysregulated hypoxia-induced autophagy promotes cancer cell survival, metabolism, and metastasis, thereby contributing to treatment resistance. Targeting HIF-mediated pathways or modulating autophagic processes offers the potential to improve traditional cancer therapies and overcome drug resistance. Pharmacological inhibitors of HIFs or autophagy, either alone or in combination with other treatments, may disrupt the pro-survival mechanisms within the hypoxic tumor microenvironment. Further research is needed to elucidate the intricate interplay between HIF signaling and the autophagy machinery in cancer cells. Understanding these processes could pave the way for novel therapeutic strategies to enhance treatment outcomes and combat drug resistance. This review highlights the complex relationship between HIFs and autophagy in cancer development and therapy, offering insights into how targeting these pathways may improve patient outcomes.
Collapse
Affiliation(s)
- Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| |
Collapse
|
31
|
Trisal A, Singh AK. Mechanisms and early efficacy data of caloric restriction and caloric restriction mimetics in neurodegenerative disease. Neuroscience 2025; 567:235-248. [PMID: 39761825 DOI: 10.1016/j.neuroscience.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
Neurodegenerative disorders (NDDs) have been prevalent for more than a decade, and the number of individuals affected per year has increased exponentially. Among these NDDs, Alzheimer's disease, which causes extreme cognitive impairment, and Parkinson's disease, characterized by impairments in motor activity, are the most prevalent. While few treatments are available for clinical practice, they have minimal effects on reversing the neurodegeneration associated with these debilitating diseases. Lifestyle modifications and dietary choices are emerging and promising approaches to combat these disorders. Of the lifestyle changes that one could adopt, a major habit is caloric restriction. Caloric restriction (CR) is a lifestyle modification in which the amount of calories ingested is reduced to a significant amount without resulting in malnutrition. However, maintaining such a lifestyle is challenging. As alternatives, certain compounds have been recognized to mimic the effects produced by CR. These compounds are called caloric restriction mimetics (CRMs). Among these compounds, some have been designated established CRMs, namely, resveratrol, metformin, and rapamycin, whereas several other candidates are termed potential CRMs because of a lack of conclusive evidence of their effects. The potential CRMs discussed in this review are quercetin, chrysin, astragalin, apigenin, curcumin, epigallocatechin-3-gallate, and NAD+ precursors. This review aims to provide an overview of these CRMs' effectiveness in preventing neurodegenerative disorders associated with aging. Moreover, we highlight the clinical relevance of these compounds by discussing in detail the results of clinical trials on them.
Collapse
Affiliation(s)
- Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India; Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Abhishek Kumar Singh
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India; Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India.
| |
Collapse
|
32
|
Wu S, Zhuang H, Zhou X, Li K. NRBF2 plays a crucial role in the acquisition process of learning and memory, independent of the Vps34 complex. Front Behav Neurosci 2025; 19:1529522. [PMID: 40013119 PMCID: PMC11861080 DOI: 10.3389/fnbeh.2025.1529522] [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: 11/28/2024] [Accepted: 01/29/2025] [Indexed: 02/28/2025] Open
Abstract
Introduction NRBF2, a component of autophagy-associated PIK3C3/VPS34-containing phosphatidylinositol 3-kinase complex, plays a crucial role in learning and memory processes, yet its specific impact on memory and the underlying molecular mechanisms remains unclear. Methods Here, we utilized NRBF2 knockout mice to examine its influence on the time course of fear memory. Employing quantitative PCR, Western blot analysis, behavioral tests, and electrophysiology, we investigated the mechanisms through which NRBF2 affects memory processing. Results We observed an increase in Nrbf2 mRNA levels at 6 and 12 h, and protein levels at 6 h post fear conditioning. Depletion of NRBF2 impaired memory acquisition, short-term, and long-term memory without causing any anxiety-like behavior. Interestingly, inhibition of Vps34 and autophagy by SAR405 disrupted fear memory consolidation, while leaving memory acquisition, short-term memory, and long-term potentiation (LTP) unaffected. Our results suggested that NRBF2 deletion impaired memory acquisition through an autophagy-independent pathway and provided novel insights into the role of NRBF2 in the central nervous system. Discussion This study offer new insights into the role of NRBF2 and highlight the potential of targeting NRBF2 as a therapeutic strategy for addressing cognitive deficits associated with various disorders.
Collapse
Affiliation(s)
- Songfen Wu
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Department of Pharmacy, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Guangzhou, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Haicai Zhuang
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Department of Pharmacy, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Guangzhou, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xidan Zhou
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kuan Li
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Department of Pharmacy, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Guangzhou, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
33
|
Liu D, Wang H, Li J, Sheng S, Wang S, Tian Y. Non-lethal sonodynamic therapy mitigates hypertensive renal fibrosis through the PI3K/AKT/mTORC1-autophagy pathway. Sci Rep 2025; 15:4534. [PMID: 39915557 PMCID: PMC11802789 DOI: 10.1038/s41598-025-86973-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
Hypertension constitutes a significant public health concern, characterized by a high incidence and mortality rate. Hypertensive kidney disease is a prevalent complication associated with hypertension and is the second leading cause of end-stage renal disease (ESRD). Renal fibrosis linked to hypertension has emerged as the third leading cause of disease in dialysis patients. Autophagy activity is crucial for maintaining homeostasis, vitality, and physiological function of kidney cells, while also protecting the kidneys from fibrosis. The deficiency of autophagy will increase the sensitivity of the kidney to the damage, leading to impaired renal function, accumulation of damaged mitochondria and more severe of renal fibrosis. However, enhancing autophagy by activating the PI3K/AKT, AMPK, and mTOR pathways, improves podocyte injury and renal pathological changes, and ameliorates renal function. Current clinical interventions aimed at halting or reversing renal fibrosis in hypertensive patients are notably limited in their efficacy. Here, we present Non-lethal Sonodynamic Therapy (NL-SDT), in which ultrasound is used to activate locally sonosensitizers, thereby stimulating the production of reactive oxygen species for the purpose of modulating cell function or fate, as a novel methodology to inhibit progression of hypertensive renal fibrosis.To confirm whether NL-SDT can reduce hypertensive renal fibrosis and its mechanism. The mice model of hypertensive renal fibrosis was established by using osmotic minipumps (Alzet model 2004, Cupertino, CA) equipped with angiotensin-II (Ang II). The pumps were implanted in mice, ensuring constant infusion of Ang II at a dose of 1.0 µg/kg per minute for 4 weeks. The mice were exposed to 0.4 W/cm2 intensity ultrasonic radiation for 15 min at 4 h post injection of sinoporphyrin sodium (DVDMS) (4 mg/kg) into the caudal vein was repeated weekly for 4 treatments. The kidney from mice was stained with masson's trichrome staining for collagen fiber expression, while alpha-smooth muscle actin (α-SMA) expression was determined via immunohistochemical staining. The protein levels of fibrosis parameters (α-SMA, collagen I, vimentin), pathway-related proteins (PI3K, AKT, mTORC1) and autophagy-related protein LC3B were determined using western blotting. Intracellular reactive oxygen species (ROS) levels were detected using DCFH-DA probe. Immunofluorescence was also used to observe the expression of α-SMA and E-cadherin in cells. Pathway-related protein inhibitors (the autophagy-related inhibitor 3-methyladenine (3-MA), chloroquine (CQ), ROS inhibitor N-acetyl-L-cysteine (NAC) were applied, and autophagosome changes were observed under transmission electron microscopy. Immunofluorescence was used to observe LC3 spot formation within cells.We obtained the following results via animal and cellular research. In vivo, (1) The collagen area of renal tissue was increased significantly in Ang II group (50.6%). The positive expression of α-SMA was increased significantly (37.8%). (2) The collagen area decreased after NL-SDT treatment (34.8%). The expression of α-SMA was decreased too (48.9%). The expression of LC3B increased in NL-SDT group. (3) The effect of NL-SDT on reducing renal fibrosis can be changed by rapamycin and CQ. In vitro. (1) The expression of α-SMA, collagen I and vimentin were increased significantly in TGF-β1-induced NRK-52E cells. (2) The increase of autophagosomes was observed in TGF-β1-induced NRK-52E cells after NL-SDT. The levels of ROS were increased after NL-SDT (24.8%). The effect of NL-SDT on autophagy was reversed after administration of NAC. The expression of PI3K, P-AKT and P-mTORC1 was decreased in TGF-β1-induced NRK-52E cells after NL-SDT. NL-SDT inhibited the transition of epithelial cells into myofibroblasts by activating PI3K-AKT-mTORC1-autophagy pathway in TGF-β1-induced NRK-52E cells. (3) The administration of the pathway inhibitors showed a reciprocal effect on NL-SDT-inhibited epithelial-mesenchymal transition (EMT).(1) NL-SDT reduced blood pressure temporarily in mice model of hypertensive renal fibrosis induced by Ang II. (2) NL-SDT alleviated renal fibrosis in mice model of hypertensive renal fibrosis induced by Ang II. (3) NL-SDT promoted autophagy by inhibiting PI3K-AKT-mTORC1 signaling pathway and alleviated renal fibrosis in mice model of hypertensive renal fibrosis induced by Ang II. NL-SDT is a non-invasive and efficacious regimen to inhibit renal fibrosis. It may be a new approach for clinical treatment of renal fibrosis, delaying or reducing the occurrence of ESRD.
Collapse
Affiliation(s)
- DanDan Liu
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China
| | - Hui Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China
| | - Jialong Li
- Department of Pathophysiology and Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, 150086, PR China
| | - Siqi Sheng
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China
| | - Shu Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China.
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China.
| |
Collapse
|
34
|
Li C, Yuan Y, Jia Y, Zhou Q, Wang Q, Jiang X. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies. Front Immunol 2025; 16:1534263. [PMID: 39963130 PMCID: PMC11830604 DOI: 10.3389/fimmu.2025.1534263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cellular aging is a multifactorial and intricately regulated physiological process with profound implications. The interaction between cellular senescence and cancer is complex and multifaceted, senescence can both promote and inhibit tumor progression through various mechanisms. M6A methylation modification regulates the aging process of cells and tissues by modulating senescence-related genes. In this review, we comprehensively discuss the characteristics of cellular senescence, the signaling pathways regulating senescence, the biomarkers of senescence, and the mechanisms of anti-senescence drugs. Notably, this review also delves into the complex interactions between senescence and cancer, emphasizing the dual role of the senescent microenvironment in tumor initiation, progression, and treatment. Finally, we thoroughly explore the function and mechanism of m6A methylation modification in cellular senescence, revealing its critical role in regulating gene expression and maintaining cellular homeostasis. In conclusion, this review provides a comprehensive perspective on the molecular mechanisms and biological significance of cellular senescence and offers new insights for the development of anti-senescence strategies.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Yixiao Yuan
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - YingDong Jia
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Zhou
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Xiulin Jiang
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
35
|
Bao Y, Ma Y, Huang W, Bai Y, Gao S, Xiu L, Xie Y, Wan X, Shan S, Chen C, Qu L. Regulation of autophagy and cellular signaling through non-histone protein methylation. Int J Biol Macromol 2025; 291:139057. [PMID: 39710032 DOI: 10.1016/j.ijbiomac.2024.139057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Autophagy is a highly conserved catabolic pathway that is precisely regulated and plays a significant role in maintaining cellular metabolic balance and intracellular homeostasis. Abnormal autophagy is directly linked to the development of various diseases, particularly immune disorders, neurodegenerative conditions, and tumors. The precise regulation of proteins is crucial for proper cellular function, and post-translational modifications (PTMs) are key epigenetic mechanisms in the regulation of numerous biological processes. Multiple proteins undergo PTMs that influence autophagy regulation. Methylation modifications on non-histone lysine and arginine residues have been identified as common PTMs critical to various life processes. This paper focused on the regulatory effects of non-histone methylation modifications on autophagy, summarizing related research on signaling pathways involved in autophagy-related non-histone methylation, and discussing current challenges and clinical significance. Our review concludes that non-histone methylation plays a pivotal role in the regulation of autophagy and its associated signaling pathways. Targeting non-histone methylation offers a promising strategy for therapeutic interventions in diseases related to autophagy dysfunction, such as cancer and neurodegenerative disorders. These findings provide a theoretical basis for the development of non-histone-methylation-targeted drugs for clinical use.
Collapse
Affiliation(s)
- Yongfen Bao
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China
| | - Yaoyao Ma
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China
| | - Wentao Huang
- Department of Physiology, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Yujie Bai
- Department of Scientific Research and Education, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, China
| | - Siying Gao
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Luyao Xiu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuyang Xie
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinrong Wan
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shigang Shan
- School of Public Health and Nursing, Hubei University of Science and Technology, Hubei 437000, China
| | - Chao Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lihua Qu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China.
| |
Collapse
|
36
|
Sogame Y, Saito R, Hakozaki S. Resting Cyst Formation as a Strategy for Environmental Adaptation in Colpodid Ciliates. Zoolog Sci 2025; 42. [PMID: 39932756 DOI: 10.2108/zs240058] [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: 06/28/2024] [Accepted: 12/04/2024] [Indexed: 05/08/2025]
Abstract
Resting cyst formation is a strategic aspect of the life cycle of some eukaryotes such as protists, and particularly ciliates, that enables adaptation to unfavorable environmental conditions. The formation of resting cysts involves large scale morphological and physiological changes that provide tolerance of extreme environmental stresses. The resting cyst shows suppression of normal features of life such as eating, moving, proliferation, and even mitochondrial metabolic activity, and appears lifeless. This review discusses resting cyst formation in the ciliates Colpoda as a representative model of cyst-forming organisms, and focusses on morphogenesis, molecular events, tolerances, and metabolic activities in resting cysts.
Collapse
Affiliation(s)
- Yoichiro Sogame
- Department of Applied Chemistry and Biochemistry, National Institute of Technology, Fukushima College, Iwaki, Fukushima 970-8034, Japan,
| | - Ryota Saito
- Department of Applied Chemistry and Biochemistry, National Institute of Technology, Fukushima College, Iwaki, Fukushima 970-8034, Japan
| | - Shuntaro Hakozaki
- Department of Applied Chemistry and Biochemistry, National Institute of Technology, Fukushima College, Iwaki, Fukushima 970-8034, Japan
| |
Collapse
|
37
|
Zhang Y, Zhang N, Zhang Y, Li Y, Yang N, Cai Y, Tan C, Zhao J, Li W, Liu Y, Rui X, Wu J, Fu Y, Liu G. Potassium molybdate blocks APN-dependent coronavirus entry by degrading receptor via PIK3C3-mediated autophagy. J Virol 2025; 99:e0144924. [PMID: 39641621 PMCID: PMC11784013 DOI: 10.1128/jvi.01449-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Swine enteric coronaviruses pose a significant challenge to the global pig industry, inflicting severe diarrhea and high mortality rates among piglets, and resulting in substantial economic losses. In our clinical practice, we observed that the addition of potassium molybdate (PM) to the feed could dramatically reduce diarrhea and diarrhea-related mortality in piglets. However, the underlying mechanisms remain elusive and merit further investigation. In this study, we revealed that PM effectively inhibited the infection of both aminopeptidase N (APN)-dependent coronaviruses, transmissible gastroenteritis virus (TGEV), and porcine respiratory coronavirus (PRCV), both in vitro and ex vivo. Specifically, PM was found to block TGEV and PRCV penetration by degrading the cell receptor APN through the upregulation of phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3) expression. In addition, knockdown and knockout of PIK3C3 resulted in the attenuation of PM-induced autophagy, thereby rescuing APN expression and viral infection. Correspondingly, replenishment of PIK3C3 in PIK3C3-null ST cells restored PM-mediated APN degradation and successfully blocked viral entry. Furthermore, our findings demonstrated that PM promoted the assembly of the PIK3C3-BECN1-ATG14 complex, leading to induced autophagic degradation by upregulating PIK3C3 Ser249 phosphorylation. In vivo experiments further confirmed that PM-induced PIK3C3-mediated autophagic degradation of APN, thereby limiting the pathogenicity of TGEV. In summary, our study for the first time identified the mechanism by which PM blocked TGEV and PRCV internalization by degrading the cell receptor APN via PIK3C3-mediated autophagy. This study provides valuable insights and potential strategies for preventing APN-restricted coronavirus infection.IMPORTANCEAminopeptidase N (APN) is one of the most important host receptors of coronavirus. Modulating APN expression can represent a novel approach for controlling APN-dependent coronaviruses and their variants infection. Here we found that a chemical compound potassium molybdate (PM) negatively regulates APN expression by inducing phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3)-mediated autophagy against APN-dependent coronavirus internalization, including transmissible gastroenteritis virus (TGEV) and porcine respiratory coronavirus (PRCV). Furthermore, PM can promote PIK3C3-BECN1-ATG14 complex assembly to induce autophagic degradation of APN by upregulating PIK3C3 Ser249 phosphorylation. Lastly, results from pig experiments also confirmed that PM can trigger PIK3C3-mediated autophagic degradation of APN to restrict TGEV pathogenicity in vivo without toxicity. Our findings underscore the promising potential of PM as an effective agent against APN-dependent coronavirus and potentially emerging viral disease entry.
Collapse
Affiliation(s)
- Yunhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liege, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Na Zhang
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yue Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ning Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liege, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yifei Cai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- Nutritional Biology, Wageningen University and Research, Wageningen, Netherlands
| | - Chen Tan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Molecular and Cellular Epigenetics (GIGA) and Molecular Biology (TERRA), University of Liege, Liege, Belgium
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Jing Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wenjie Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuanyuan Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Xue Rui
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Junfei Wu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuguang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Guangliang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute Chinese Academy of Agricultural Sciences, Lanzhou, China
- Hainan Key Laboratory of Tropical Animal Breeding and Infectious Disease Research, Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| |
Collapse
|
38
|
Zhang S, Hu W, Tang Y, Chen X. Identification and validation of key autophagy-related genes in lupus nephritis by bioinformatics and machine learning. PLoS One 2025; 20:e0318280. [PMID: 39869603 PMCID: PMC11771862 DOI: 10.1371/journal.pone.0318280] [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: 08/19/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025] Open
Abstract
INTRODUCTION Lupus nephritis (LN) is one of the most frequent and serious organic manifestations of systemic lupus erythematosus (SLE). Autophagy, a new form of programmed cell death, has been implicated in a variety of renal diseases, but the relationship between autophagy and LN remains unelucidated. METHODS We analyzed differentially expressed genes (DEGs) in kidney tissues from 14 LN patients and 7 normal controls using the GSE112943 dataset. Key modules and their contained genes were identified utilizing weighted gene co-expression network analysis (WGCNA). Differentially expressed autophagy-related genes (DE-ARGs) among DEGs, key module genes and autophagy-related genes (ARGs) were obtained by venn plot, and subjected to protein-protein interaction network construction. Two machine learning methods were applied to identify signature genes. The area under the receiver operating characteristic (ROC) curves was used to assess the accuracy of the signature genes. We also analyzed immune cell infiltration in LN. Additionally, the association between key genes and kidney diseases was predicted. Finally, key genes expression in kidney was verified by clinical samples and animal experiments. RESULTS A total of 10304 DEGs were identified in GSE1129943 and 29 modules were identified in WGCNA. Among them, the brown module and coral 2 module exhibited significant correlation with LN (cor = 0.86, -0.84, p<0.001). Machine learning techniques identified 5 signature genes, but only 2 were validated in the external dataset GSE32591, namely MAP1LC3B (AUC = 0.920) and TNFSF10 (AUC = 0.937), which are involved in autophagy and apoptosis. Immune infiltration analysis suggested that these key genes may be associated with immune cell infiltration in LN. In addition, these genes have been linked to a variety of renal diseases, and their expression was verified in kidney tissues in LN patients and lupus mice. CONCLUSION MAP1LC3B and TNFSF10 may be key autophagy-related genes in LN. These key genes have the potential to provide new insights into the molecular diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Su Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Yelin Tang
- General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Xiaoqing Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| |
Collapse
|
39
|
Wan K, Li J, Ma L, Chen T, Chen Y, Li Z, Zouboulis CC, Wang GL, Wang J. Camellia saponin modulates oleic acid/linoleic acid-induced lipogenesis in human sebocytes through lipophagy activation. Int J Cosmet Sci 2025. [PMID: 39844373 DOI: 10.1111/ics.13047] [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: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Oily skin not only threatens people with aesthetic and hygienic discomfort but also confronts them with annoying skin problems. To explore new skin care ingredients from herbal or plant extracts and understand their underlying mechanism for sebum control would assist in the discovery of desirable sebosuppressive agents, though it is still a deserving and challenging task. AIM To explore the effect of Camellia saponin (CS) on modulating the lipogenesis of human sebocytes. Moreover, to explore the underlying mechanism of CS on oleic acid/linoleic acid (OL) mixture stimulated lipid accumulation. METHODS The lipid accumulation model of cells was constructed by OL-induction in vitro. The lipid synthesis in SZ95 sebocytes was detected by Oil Red O, Nile Red and BODIPY staining and the distribution of lipid droplets and autophagosomes were evaluated by transmission electron microscopy (TEM). Fluorescence staining, immunofluorescence and western blot (WB) were used to characterize the spatial localization of lipid droplets (LDs)/autophagosome/lysosome, the levels of LC3 and P62 proteins related to intracellular autophagy, as well as the pH of lysosome. RESULTS CS treatment significantly relieved OL-induced lipid accumulation in SZ95 sebocytes. Furthermore, CS maintained lysosomal acid environment to promote the fusion of autophagosome and lysosome, thus recovering the OL-induced blockage of autophagy flow. We also found that CS activated AMPK, and down-regulated mTOR in SZ95 sebocytes. CONCLUSION CS was able to relieve OL-stimulated sebum accumulation in cultured human SZ95 sebocytes through lipophagy, in which process CS maintained lysosomal acid environment and activated the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Kaibo Wan
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Jian Li
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Ling Ma
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Timson Chen
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Ya Chen
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Zhizhen Li
- Adolph Innovation Laboratory, Guangzhou AOGU Cosmetics Co., Ltd., Guangzhou, China
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Staedtisches Klinikum, Brandenburg Medical School Theodore Fontane, Dessau, Germany
| | - Guang-Li Wang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| | - Jing Wang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China
| |
Collapse
|
40
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat 2025; 78:101170. [PMID: 39603146 DOI: 10.1016/j.drup.2024.101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Macroautophagy/autophagy is a highly conserved evolutionary mechanism involving lysosomes for the degradation of cytoplasmic components including organelles. The constitutive, basal level of autophagy is fundamental for preserving cellular homeostasis; however, alterations in autophagy can cause disease pathogenesis, including cancer. The role of autophagy in cancer is particularly complicated, since this process acts both as a tumor suppressor in precancerous stages but facilitates tumor progression during carcinogenesis and later stages of cancer progression. This shift between anti-tumor and pro-tumor roles may be influenced by genetic and environmental factors modulating key pathways such as those involving autophagy-related proteins, the PI3K-AKT-MTOR axis, and AMPK, which often show dysregulation in tumors. Autophagy regulates various cellular functions, including metabolism of glucose, glutamine, and lipids, cell proliferation, metastasis, and several types of cell death (apoptosis, ferroptosis, necroptosis and immunogenic cell death). These multifaceted roles demonstrate the potential of autophagy to affect DNA damage repair, cell death pathways, proliferation and survival, which are critical in determining cancer cells' response to chemotherapy. Therefore, targeting autophagy pathways presents a promising strategy to combat chemoresistance, as one of the major reasons for the failure in cancer patient treatment. Furthermore, autophagy modulates immune evasion and the function of immune cells such as T cells and dendritic cells, influencing the tumor microenvironment and cancer's biological behavior. However, the therapeutic targeting of autophagy is complex due to its dual role in promoting survival and inducing cell death in cancer cells, highlighting the need for strategies that consider both the beneficial and detrimental effects of autophagy modulation in cancer therapy. Hence, both inducers and inhibitors of autophagy have been introduced for the treatment of cancer. This review emphasizes the intricate interplay between autophagy, tumor biology, and immune responses, offering insights into potential therapeutic approaches that deploy autophagy in the cancer suppression.
Collapse
Affiliation(s)
- Xuegang Niu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi You
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Kaijian Hou
- School of Public Health(Long Hu people hospital), Shantou University, Shantou, 515000, Guangdong, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL 60532, USA
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bin Gao
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Lingzhi Wang
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Pfuhler L, Awad S, Skipper W, Lavietes J, Sah T, Ho K, Ivanova R, Cooke A. The autophagy initiation factor ATG13 mRNA is stabilized by the RNA-binding protein YBX3. FEBS Lett 2025; 599:89-99. [PMID: 39414370 PMCID: PMC11726137 DOI: 10.1002/1873-3468.15035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/20/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
Autophagy, a highly conserved form of cellular recycling, is essential for cellular homeostasis. Its dysregulation has been linked to neurodegenerative diseases and various cancers, including breast cancer. We set out to determine if the RNA-binding protein (RBP) YBX3 regulates autophagy mRNAs, as previous findings suggest YBX3 depletion reduces distinct autophagy transcripts. We found that YBX3 interacts with and stabilizes the mRNA of the autophagy initiation factor ATG13 in HeLa, which in turn increases ATG13 protein expression. We have shown that this requires the 3' untranslated region (UTR) of ATG13 and occurs in other human cell lines, including HEK293, HepG2, and HCT116. Together, our data suggest a novel regulatory role for YBX3 of autophagy initiation through posttranscriptional control of ATG13 mRNA stability.
Collapse
Affiliation(s)
- Liva Pfuhler
- Haverford CollegePAUSA
- Present address:
University of Pittsburgh School of MedicinePittsburghPAUSA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Huang J, Wang J. Selective protein degradation through chaperone‑mediated autophagy: Implications for cellular homeostasis and disease (Review). Mol Med Rep 2025; 31:13. [PMID: 39513615 PMCID: PMC11542157 DOI: 10.3892/mmr.2024.13378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/03/2024] [Indexed: 11/15/2024] Open
Abstract
Cells rely on autophagy for the degradation and recycling of damaged proteins and organelles. Chaperone-mediated autophagy (CMA) is a selective process targeting proteins for degradation through the coordinated function of molecular chaperones and the lysosome‑associated membrane protein‑2A receptor (LAMP2A), pivotal in various cellular processes from signal transduction to the modulation of cellular responses under stress. In the present review, the intricate regulatory mechanisms of CMA were elucidated through multiple signaling pathways such as retinoic acid receptor (RAR)α, AMP‑activated protein kinase (AMPK), p38‑TEEB‑NLRP3, calcium signaling‑NFAT and PI3K/AKT, thereby expanding the current understanding of CMA regulation. A comprehensive exploration of CMA's versatile roles in cellular physiology were further provided, including its involvement in maintaining protein homeostasis, regulating ferroptosis, modulating metabolic diversity and influencing cell cycle and proliferation. Additionally, the impact of CMA on disease progression and therapeutic outcomes were highlighted, encompassing neurodegenerative disorders, cancer and various organ‑specific diseases. Therapeutic strategies targeting CMA, such as drug development and gene therapy were also proposed, providing valuable directions for future clinical research. By integrating recent research findings, the present review aimed to enhance the current understanding of cellular homeostasis processes and emphasize the potential of targeting CMA in therapeutic strategies for diseases marked by CMA dysfunction.
Collapse
Affiliation(s)
- Jiahui Huang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- College of Traditional Chinese Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Jiazhen Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
43
|
Xu S, Wang Z, Guo F, Zhang Y, Peng H, Zhang H, Liu Z, Cao C, Xin G, Chen YY, Fu J. Mitophagy in ischemic heart disease: molecular mechanisms and clinical management. Cell Death Dis 2024; 15:934. [PMID: 39737905 DOI: 10.1038/s41419-024-07303-3] [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: 08/14/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025]
Abstract
The influence of the mitochondrial control system on ischemic heart disease has become a major focus of current research. Mitophagy, as a very crucial part of the mitochondrial control system, plays a special role in ischemic heart disease, unlike mitochondrial dynamics. The published reviews have not explored in detail the unique function of mitophagy in ischemic heart disease, therefore, the aim of this paper is to summarize how mitophagy regulates the progression of ischemic heart disease. We conclude that mitophagy affects ischemic heart disease by promoting cardiomyocyte hypertrophy and fibrosis, the progression of oxidative stress, the development of inflammation, and cardiomyocyte death, and that the specific mechanisms of mitophagy are worthy of further investigation.
Collapse
Affiliation(s)
- Shujuan Xu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110122, China
| | - Fan Guo
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yehao Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Han Peng
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Huiyu Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zixin Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Ce Cao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Gaojie Xin
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yuan Yuan Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jianhua Fu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
44
|
Kapuy O, Holczer M, Csabai L, Korcsmáros T. Oscillatory autophagy induction is enabled by an updated AMPK-ULK1 regulatory wiring. PLoS One 2024; 19:e0313302. [PMID: 39724154 DOI: 10.1371/journal.pone.0313302] [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: 07/09/2024] [Accepted: 10/23/2024] [Indexed: 12/28/2024] Open
Abstract
Autophagy-dependent survival relies on a crucial oscillatory response during cellular stress. Although oscillatory behaviour is typically associated with processes like the cell cycle or circadian rhythm, emerging experimental and theoretical evidence suggests that such periodic dynamics may explain conflicting experimental results in autophagy research. In this study, we demonstrate that oscillatory behaviour in the regulation of the non-selective, stress-induced macroautophagy arises from a series of interlinked negative and positive feedback loops within the mTORC1-AMPK-ULK1 regulatory triangle. While many of these interactions have been known for decades, recent discoveries have revealed how mTORC1, AMPK, and ULK1 are truly interconnected. Although these new findings initially appeared contradictory to established models, additional experiments and our systems biology analysis clarify the updated regulatory structure. Through computational modelling of the autophagy oscillatory response, we show how this regulatory network governs autophagy induction. Our results not only reconcile previous conflicting experimental observations but also offer insights for refining autophagy regulation and advancing understanding of its mechanisms of action.
Collapse
Affiliation(s)
- Orsolya Kapuy
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Marianna Holczer
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Luca Csabai
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Tamás Korcsmáros
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Quadram Institute, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
45
|
Brokowska J, Herman-Antosiewicz A, Hać A. Isothiocyanates induce autophagy and inhibit protein synthesis in primary cells via modulation of AMPK-mTORC1-S6K1 signaling pathway, and protect against mutant huntingtin aggregation. Eur J Nutr 2024; 64:46. [PMID: 39680190 PMCID: PMC11649724 DOI: 10.1007/s00394-024-03539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 11/04/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Autophagy is a degradation process whose activation underlies beneficial effects of caloric restriction. Isothiocyanates (ITCs) induce autophagy in cancer cells, however, their impact on primary cells remains insufficiently explored, particularly in non-epithelial cells. The aim of this study was to investigate whether ITCs induce autophagy in primary (non-immortalized) mesenchymal cells and if so, to determine the molecular mechanism underlying its activation and consequences on cell functioning. METHODS Primary human dermal fibroblasts (HDFa) and prostate cancer cells (PC3) as well as two ITCs, sulforaphane and phenethyl isothiocyanate, were applied. Cell viability was measured by the MTT test, protein synthesis - by 3H-leucine incorporation, and protein level - by immunoblotting. A number of mutant huntingtin (mHtt) aggregates was assessed by fluorescence microscopy. RESULTS Both ITCs efficiently induced autophagy in fibroblasts which coincided with suppression of mTORC1 - a negative autophagy regulator - and protein synthesis arrest. A dephosphorylation of mTORC1 substrate, S6K1, and ribosomal S6 protein was preceded by activation of AMPK, an inhibitor of mTORC1 and autophagy activator. A similar response was observed in phenethyl isothiocyanate-treated prostate cancer cells. We also showed that ITCs-induced autophagy and/or translation block do not affect cells viability and can protect cells against an accumulation of mHtt aggregates - a main cause of Huntington's disease. CONCLUSION Our study showed that ITCs induce autophagy and inhibit protein synthesis in both primary mesenchymal and cancer cells via modulation of the AMPK-mTORC1-S6K1 pathway. Moreover, it suggests that ITCs might have a potential in developing therapeutics for Huntington's disease.
Collapse
Affiliation(s)
- Joanna Brokowska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk, 80-308, Poland
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Anna Herman-Antosiewicz
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk, 80-308, Poland
| | - Aleksandra Hać
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk, 80-308, Poland.
| |
Collapse
|
46
|
Hong J, Liu W, Xiao X, Gajendran B, Ben-David Y. Targeting pivotal amino acids metabolism for treatment of leukemia. Heliyon 2024; 10:e40492. [PMID: 39654725 PMCID: PMC11626780 DOI: 10.1016/j.heliyon.2024.e40492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
Metabolic reprogramming is a crucial characteristic of cancer, allowing cancer cells to acquire metabolic properties that support their survival, immune evasion, and uncontrolled proliferation. Consequently, targeting cancer metabolism has become an essential therapeutic strategy. Abnormal amino acid metabolism is not only a key aspect of metabolic reprogramming but also plays a significant role in chemotherapy resistance and immune evasion, particularly in leukemia. Changes in amino acid metabolism in tumor cells are typically driven by a combination of signaling pathways and transcription factors. Current approaches to targeting amino acid metabolism in leukemia include inhibiting amino acid transporters, blocking amino acid biosynthesis, and depleting specific amino acids to induce apoptosis in leukemic cells. Different types of leukemic cells rely on the exogenous supply of specific amino acids, such as asparagine, glutamine, arginine, and tryptophan. Therefore, disrupting the supply of these amino acids may represent a vulnerability in leukemia. This review focuses on the pivotal role of amino acids in leukemia metabolism, their impact on leukemic stem cells, and their therapeutic potential.
Collapse
Affiliation(s)
- Jiankun Hong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| | - Babu Gajendran
- Institute of Pharmacology and Biological Activity, Natural Products Research Center of Guizhou Province, Guiyang, Guizhou, 550014, PR China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou Province, PR China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guian New Disctrict, 561113, Guizhou, PR China
- Natural Products Research Center of Guizhou. PR China
| |
Collapse
|
47
|
Galhuber M, Thedieck K. ODE-based models of signaling networks in autophagy. CURRENT OPINION IN SYSTEMS BIOLOGY 2024; 39:100519. [DOI: 10.1016/j.coisb.2024.100519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
48
|
Fakhredini F, Alidadi H, Mahdavinia M, Khorsandi L. Morin promotes autophagy in human PC3 prostate cancer cells by modulating AMPK/mTOR/ULK1 signaling pathway. Tissue Cell 2024; 91:102557. [PMID: 39265522 DOI: 10.1016/j.tice.2024.102557] [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: 06/23/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
AMP-activated protein kinase (AMPK) suppresses tumorigenesis by modulating autophagy and apoptosis. This study evaluated the impact of Morin on PC3 prostate cancerous cells by examining the AMPK/ mechanistic target of rapamycin (mTOR)/ ULK1 (UNC-51-like kinase 1) pathway and autophagy process. The PC3 cells were treated with Morin (50 µg/ml) and AICAR (an AMPK activator). Cell viability, apoptosis, autophagy, and level of phosphorylated and non-phosphorylated ULK1, AMPK, and mTOR, as well as LC3B/LC3A, have been investigated. Through DAPI staining, measurement of Bax/Bcl-2 ratio, Caspase activity, and Annexin V/PI method, it has been revealed that Morin induces apoptosis and reduces the growth of PC3 cells. Morin enhanced the protein level of phosphorylated AMPK (p-AMPK) and ULK1 (p-ULK1) and decreased the expression of phosphorylated mTOR (p-mTOR) in the PC3 cells. Morin could also increase the LC3B/LC3A ratio, Acridine Orange-positive cells, expression of Beclin-1 and ATG5 genes, and decrease the p62 protein level indicating autophagy-inducing. AICAR (an AMPK activator) enhanced the impact of Morin on apoptosis, cell growth, and expression of LC3B, p-AMPK, p-ULK1, and p-mTOR proteins in the PC3 cells. These findings suggest that Morin induces apoptotic and autophagic cell death by activating AMPK and ULK1 and suppressing mTOR pathways.
Collapse
Affiliation(s)
- Fereshtesadat Fakhredini
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadis Alidadi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
49
|
Li Z, Zhang Y, Lei J, Wu Y. Autophagy in oral cancer: Promises and challenges (Review). Int J Mol Med 2024; 54:116. [PMID: 39422076 PMCID: PMC11518578 DOI: 10.3892/ijmm.2024.5440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Autophagy captures damaged or dysfunctional proteins and organelles through the lysosomal pathway to achieve proper cellular homeostasis. Autophagy possesses distinct characteristics and is given recognized functions in numerous physiological and pathological conditions, such as cancer. Early stage cancer development can be stopped by autophagy. After tumor cells have successfully undergone transformation and progressed to a late stage, the autophagy-mediated system of dynamic degradation and recycling will support cancer cell growth and adaptation to various cellular stress responses while preserving energy homeostasis. In the present study, the dual function that autophagy plays in various oral cancer development contexts and stages, the existing arguments for and against autophagy, and the ways in which autophagy contributes to oral cancer modifications, such as carcinogenesis, drug resistance, invasion, metastasis and self-proliferation, are reviewed. Special attention is paid to the mechanisms and functions of autophagy in oral cancer processes, and the most recent findings on the application of certain conventional drugs or natural compounds as novel agents that modulate autophagy in oral cancer are discussed. Overall, further research is needed to determine the validity and reliability of autophagy promotion and inhibition while maximizing the difficult challenge of increasing cancer suppression to improve clinical outcomes.
Collapse
Affiliation(s)
- Zhou Li
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, P.R. China
- Shanxi Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030000, P.R. China
| | - Yao Zhang
- Shanxi Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi 030000, P.R. China
| | - Jianhua Lei
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, P.R. China
| | - Yunxia Wu
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030000, P.R. China
| |
Collapse
|
50
|
Kanmani S, Song XM, Kanmani P, Wu XJ, Xiao-Di-Tan, Liu J, Wang JP, Minshall RD, Hu G. Enhancement of Autophagy in Macrophages via the p120-Catenin-Mediated mTOR Signaling Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1666-1675. [PMID: 39423222 PMCID: PMC11610512 DOI: 10.4049/jimmunol.2400189] [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: 04/04/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Autophagy serves as a critical regulator of immune responses in sepsis. Macrophages are vital constituents of both innate and adaptive immunity. In this study, we delved into the intricate role of p120-catenin (p120) in orchestrating autophagy in macrophages in response to endotoxin stimulation. Depletion of p120 effectively suppressed LPS-induced autophagy in both J774A.1 macrophages and murine bone marrow-derived macrophages. LPS not only elevated the interaction between p120 and L chain 3 (LC3) I/II but also facilitated the association of p120 with mammalian target of rapamycin (mTOR). p120 depletion in macrophages by small interfering RNA reduced LPS-induced dissociation of mTOR and Unc-51-like kinase 1 (ULK1), leading to an increase in the phosphorylation of ULK1. p120 depletion also enhanced LPS-triggered macrophage apoptosis, as evidenced by increased levels of cleaved caspase 3, 7-aminoactinomycin D staining, and TUNEL assay. Notably, inhibiting autophagy reversed the decrease in apoptosis caused by LPS stimulation in macrophages overexpressing p120. Additionally, the ablation of p120 inhibited autophagy and accentuated apoptosis in alveolar macrophages in LPS-challenged mice. Collectively, our findings strongly suggest that p120 plays a pivotal role in fostering autophagy while concurrently hindering apoptosis in macrophages, achieved through modulation of the mTOR/ULK1 signaling pathway in sepsis. This underscores the potential of targeting macrophage p120 as an innovative therapeutic avenue for treating inflammatory disorders.
Collapse
Affiliation(s)
- Suganya Kanmani
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Xue-Min Song
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- The Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Wuchang, 125 Donghu Road, Hubei Province, China
| | - Paulraj Kanmani
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Xiao-Jing Wu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Wuchang, 238 Liberation Road, Hubei Province, China
| | - Xiao-Di-Tan
- Department Pediatrics, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Jing Liu
- Department of Surgery/Cancer Center, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Ji-Ping Wang
- Departments of Statistics and Data Science, Northwestern University, Evanston, Illinois, United States of America
| | - Richard D. Minshall
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, 60612, United States of America
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, 60612, United States of America
| |
Collapse
|