1
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Jiang SM, Li XJ, Wang ZL, Chen ZW, Liu ZL, Li Q, Chen XL. Role of autophagy in rejection after solid organ transplantation: A systematic review of the literature. World J Transplant 2025; 15:103163. [DOI: 10.5500/wjt.v15.i3.103163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 04/18/2025] Open
Abstract
Organ transplantation has long been recognized as an effective treatment for end-stage organ failure, metabolic diseases, and malignant tumors. However, graft rejection caused by major histocompatibility complex mismatch remains a significant challenge. While modern immunosuppressants have made significant strides in reducing the incidence and risk of rejection, they have not been able to eliminate it completely. The intricate mechanisms underlying transplant rejection have been the subject of intense investigation by transplant immunologists. Among these factors, autophagy has emerged as a key player. Autophagy is an evolutionarily conserved mechanism in eukaryotic cells that mediates autophagocytosis and cellular protection. This process is regulated by autophagy-related genes and their encoded protein families, which maintain the material and energetic balance within cells. Additionally, autophagy has been reported to play crucial roles in the development, maturation, differentiation, and responses of immune cells. In the complex immune environment following transplantation, the role and mechanisms of autophagy are gradually being revealed. In this review, we aim to explore the current understanding of the role of autophagy in solid organ rejection after transplantation. Furthermore, we delve into the therapeutic advancements achieved by targeting autophagy involved in the rejection process.
Collapse
Affiliation(s)
- Shu-Min Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Xue-Jiao Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, China
| | - Zi-Lin Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Zhi-Wei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Zhi-Long Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Qiang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Xiao-Long Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
3
|
Zhang S, Zhang T, Cao Z, Yang Y, Lü P. Hijacking the autophagy-apoptosis crosstalk: African swine fever virus orchestrates immune evasion via host remodeling for viral pathogenesis. Microb Pathog 2025; 204:107609. [PMID: 40250498 DOI: 10.1016/j.micpath.2025.107609] [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: 10/25/2024] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
African swine fever (ASF) is an acute, highly fatal hemorrhagic disease of domestic and wild pigs caused by African swine fever virus (ASFV). ASFV, a large double-stranded DNA virus of the Asfarviridae family, is highly infectious and pathogenic. Through modulation of host apoptosis and autophagy pathways, the virus subverts innate immune surveillance to promote its replication and dissemination. Following ASFV infection, domestic pigs may exhibit 100 % morbidity and mortality rates with highly virulent strains, constituting a major threat to the global pork industry. Nowadays, ASF is listed as a notifiable terrestrial animal disease by the World Organisation for Animal Health (WOAH). Therefore, elucidating ASFV's pathogenic mechanisms, particularly its molecular regulation of apoptosis and autophagy, is crucial for developing effective ASF control and prevention strategies. This review comprehensively summarizes the pathogenic mechanisms of ASFV, with particular focus on the autophagy-apoptosis crosstalk and viral manipulation of these cellular processes. These insights not only improve our understanding of ASFV-mediated immune evasion mechanisms but also provide valuable references for developing ASF control strategies targeting apoptosis and autophagy pathways.
Collapse
Affiliation(s)
- Simeng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| | - Tiancheng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| | - Zhaoxiao Cao
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| | - Yanhua Yang
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China.
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| |
Collapse
|
4
|
Lyu Q, Kouketsu Y, Tazaki A, Kato M, Motooka Y, Toyokuni S. Terrestrial iron sulfide minerals induce distinct regulation of intracellular redox homeostasis and iron assimilation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118327. [PMID: 40381394 DOI: 10.1016/j.ecoenv.2025.118327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/12/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Repeated exposure to airborne terrestrial natural minerals may cause pneumoconiosis and lung cancer, among which iron sulfide is identified as an aggravating factor. In the biological system, iron-sulfur cluster is an inorganic cofactor that is evolutionarily conserved in all the living organisms. Whereas ferrous iron catalyzes the generation of hydroxyl radicals, sulfur is indispensable as a component of antioxidants, such as glutathione. Imbalanced redox homeostasis contributes to oxidative stress, causing ferroptosis, an iron-dependent regulated necrosis characterized by lipid peroxidation, resulting in various disorders. We undertook this study to understand the cellular regulatory mechanisms against major terrestrial minerals containing iron and sulfur from the viewpoint of cellular redox. We used fundamental iron sulfide minerals collected from natural sources to treat human macrophage and fibroblast cells and investigated the biological responses. Alterations in sulfane sulfur, glutathione and iron have been analyzed using either specific fluorescent probes or inductively coupled plasma mass spectrometry. Iron sulfide microparticles with high Fe/S ratio (pyrrhotite; Fe1-XS) induced more reactive sulfane species and glutathione, with less catalytic iron inside cells, whereas the mineral with low Fe/S ratio (pyrite; FeS2) exhibited the opposite effects. Notably both showed cytotoxicity, where pyrite caused ferroptosis but pyrrhotite led to non-ferroptotic disruption. Furthermore, assimilated cellular excess iron was secreted via CD63(+) exosome containing iron-loaded ferritin to the extracellular space with higher iron content in pyrrhotite. Our findings suggest that iron and sulfur work complementarily in maintaining intracellular redox homeostasis, which would be crucial to understand the associated pathology.
Collapse
Affiliation(s)
- Qinying Lyu
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yui Kouketsu
- Department of Earth and Planetary Sciences, Nagoya University Graduate School of Environmental Studies, Furo-cho, Chikusa, Nagoya 484-8601, Japan
| | - Akira Tazaki
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Activity of the Institute of Innovation for Future Society of Nagoya University, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Activity of the Institute of Innovation for Future Society of Nagoya University, Japan
| | - Yashiro Motooka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Center for Low Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Furo-Cho, Chikusa-ku, Nagoya 464-8603, Japan.
| |
Collapse
|
5
|
Yang H, Chen L, Jiang Z, Li L, Hu J, Chen WH. Design, synthesis and biological evaluation of Golgi-targeting anion transporters as inducers of Golgiphagy and apoptosis in cancer cells. Eur J Med Chem 2025; 290:117519. [PMID: 40117859 DOI: 10.1016/j.ejmech.2025.117519] [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: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Disruption in the homeostasis of anions within organelles in cancer cells by synthetic small-molecule anion transporters may lead to significant inhibition in the proliferation of cancer cells. However, the specific impact of anion transporters on organelles, in particular on the Golgi apparatus remains to be explored. In this study, we designed and synthesized a novel series of Golgi-targeting anion transporters composed of squaramido moiety for transporting chloride anions and benzenesulfonamido group for targeting the Golgi apparatus. These compounds were able to efficiently facilitate the transport of anions across liposomal and cellular membranes, and exhibit significant cytotoxicity toward several selected cancer cells. Among them, compound 10 was the most active in efficiently disrupting the homeostasis of chloride anions specifically within the Golgi apparatus. This disruption led to profound perturbations in the structure and function of the Golgi apparatus, and triggered Golgiphagy and further apoptosis. More importantly, compound 10 displayed potent antitumor efficacy toward HepG2 xenograft mouse models, with low toxicity and minimal adverse effects on major organs. The present findings underscore the critical role of regulating the homeostasis of chloride anions within the Golgi apparatus in triggering the Golgiphagy and apoptosis of cancer cells, and thus provide a new strategy for the discovery of innovative chemotherapy for cancers.
Collapse
Affiliation(s)
- Haodong Yang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Li Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Zixing Jiang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Lanqing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Jinhui Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China.
| |
Collapse
|
6
|
Zamanian MY, Kamran Z, Tavakoli MR, Oghenemaro EF, Abohassan M, Kubaev A, Nathiya D, Kaur P, Zwamel AH, Abdulamer RS. The Role of ΔFosB in the Pathogenesis of Levodopa-Induced Dyskinesia: Mechanisms and Therapeutic Strategies. Mol Neurobiol 2025; 62:7393-7412. [PMID: 39890697 DOI: 10.1007/s12035-025-04720-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Levodopa-induced dyskinesia (LID) represents a significant complication associated with the long-term administration of levodopa (L-DOPA) for the treatment of Parkinson's disease (PD). This review examines the critical role of ΔFosB, a transcription factor, in the pathogenesis of LID and explores potential therapeutic interventions. ΔFosB accumulates within the striatum in response to chronic dopaminergic stimulation, thereby driving maladaptive changes that culminate in dyskinesia. Its persistent expression modifies gene transcription, influencing neuronal plasticity and contributing to the sustained presence of dyskinetic movements. This study explains how ΔFosB functions at the molecular level, focusing on its connections with dopamine D1 receptors, the cAMP/PKA signaling pathway, and its regulatory effects on downstream targets such as DARPP-32 and GluA1 AMPA receptor subunits. Additionally, it examines how neuronal nitric oxide synthase (nNOS) affects ΔFosB levels and the development of LID. This review also considers the interactions between ΔFosB and other signaling pathways, such as ERK and mTOR, in the context of LID and striatal plasticity. Emerging therapeutic strategies targeting ΔFosB and its associated pathways include pharmacological interventions like ranitidine, 5-hydroxytryptophan, and carnosic acid. Furthermore, this study addresses the role of JunD, another component of the AP-1 transcription factor complex, in the pathogenesis of LID. Understanding the molecular mechanisms by which ΔFosB contributes to LID offers promising avenues for developing novel treatments that could mitigate dyskinesia and improve the quality of life for PD patients undergoing long-term L-DOPA therapy.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Zahra Kamran
- Department of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marziye Ranjbar Tavakoli
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology & Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Resan Shakir Abdulamer
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
7
|
Liu M, Wang Y, Ren F, Zhang W, Zheng H, Shi Q, Zhang R, Gao C, Luo L, Gu J, Nie C. Alterations of retinal autophagy after a blast simulated microgravity in rats. Exp Eye Res 2025; 255:110366. [PMID: 40180275 DOI: 10.1016/j.exer.2025.110366] [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: 09/18/2024] [Revised: 02/11/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Emerging research has confirmed the crucial role of autophagy, an endogenous repair mechanism, in various blast injuries. However, its role in explosive ocular injury (EOI) under microgravity (MG) and normal gravity (NG) environments remains poorly understood. Therefore, this study aimed to investigate the changes in retinal lesions and retinal autophagy over time following EOI under both NG and MG environments. This study employed the hind-limb unloading model in Sprague-Dawley (SD) rats to simulate MG conditions and used self-made device with compressed gas to induce EOI. SD rats were randomly divided into six groups as follows: normal gravity control group (NG + non-EOI group), normal gravity model group at 1 day post-EOI injury (NG + EOI 1dpi group, n = 20), normal gravity model group at 7 days post-EOI injury (NG + EOI 7dpi group, n = 20), microgravity control group (MG + non-EOI group), microgravity model group at 1 day post-EOI injury (MG + EOI 1dpi group, n = 20), and microgravity model group at 7 days post-EOI injury (MG + EOI 7dpi group, n = 20). Evaluations of ocular health (gross pathology and histology), and retinal autophagy (histology and WB) were conducted before EOI, as well as at 1 and 7 days following EOI. Compared to the NG + non-EOI group, the NG + EOI group rats exhibited significant increases in autophagy-related proteins and genes in the retina, including Beclin1, LC3Ⅱ/LC3Ⅰ, ATF4, GRP78, CHOP, ATG5, and ATG7, along with a decrease in p62, indicating an elevation in retinal autophagy and ER-phagy levels. Retinal lesions, disintegration, and autophagosomes in the ganglion cell layer (GCL) and photoreceptor inner/outer segment layers (PISL/POSL) diminished over time in the NG + EOI group rats. Meanwhile, the MG + EOI group rats exhibited more severe retinal lesions and disintegration, along with an increased number of autophagosomes in the GCL and PISL/POSL, with these symptoms worsening over time compared to the MG + non-EOI group. Compared to the MG + non-EOI group, the MG + EOI group rats exhibited significant decreases in autophagy-related proteins and genes in the retina, including Beclin1, LC3Ⅱ/LC3Ⅰ, ATF4, GRP78, CHOP, ATG5, and ATG7, along with an increase in p62, suggesting a reduction in retinal autophagy levels. Taken together, retinal autophagy and ER-phagy may serve as a self-protective mechanism following EOI under NG conditions. However, under MG conditions, EOI may disrupt this protective mechanism, potentially causing irreversible retinal damage and increasing the risk of blindness in astronauts.
Collapse
Affiliation(s)
- Meng Liu
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuyu Wang
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Ren
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Wenqian Zhang
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Hanwen Zheng
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Quanxing Shi
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Rong Zhang
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Caiyun Gao
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Ling Luo
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Jianwen Gu
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China.
| | - Chuang Nie
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China.
| |
Collapse
|
8
|
Sun C, He W, Wang L, Hao T, Yang X, Feng W, Wu Y, Meng C, Wang Z, Chen X, Sun M, Zheng F, Zhang B. Studies on the Role of MAP4K2, SPI1, and CTSD in Osteoporosis. Cell Biochem Biophys 2025; 83:2115-2126. [PMID: 39586961 PMCID: PMC12089232 DOI: 10.1007/s12013-024-01621-1] [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] [Accepted: 11/10/2024] [Indexed: 11/27/2024]
Abstract
Osteoporosis (OP) is a prevalent skeletal disorder characterized by an imbalance between bone resorption and bone formation, resulting in a significant global burden. Previous research utilizing bioinformatics analysis has identified MAP4K2, SPI1, and CTSD as hub genes associated with OP. In this current investigation, we have successfully established a differential expression system of MAP4K2, SPI1, and CTSD in rat bone marrow mesenchymal stem cells (BMSCs) through transfection techniques. Additionally, the CCK-8 assay was employed to assess cell proliferation, while the alkaline phosphatase (ALP) activity assay and ALP staining assay were utilized to evaluate osteogenic differentiation. Alizarin red staining was employed to detect mineralization of BMSCs. Furthermore, the expression of relevant genes and molecules associated with the MAPK signaling pathway, autophagy, and apoptosis in the sera of rat BMSCs were examined using quantitative real-time polymerase chain reaction (qRT-PCR). The purpose of this study was to preliminarily investigate whether MAP4K2, SPI1, and CTSD have an effect on the osteogenic capacity of rat BMSCs and whether these genes, when differentially expressed, affect the expression of related genes in the MAPK, autophagy, and apoptosis signaling pathways and thus the osteogenic function of BMSCs. In summary, the findings of this study indicate that MAP4K2 and CTSD exert significant influence on the proliferation, osteogenic differentiation, and mineralization processes of rat BMSCs cells. Furthermore, these proteins may contribute to the development of OP through their involvement in the regulation of autophagy and apoptosis. Conversely, our investigation did not reveal any discernible impact of SPI1 on OP-related phenotypes. Consequently, this research serves as a fundamental basis for further exploration of potential therapeutic targets for the treatment of OP.
Collapse
Affiliation(s)
- Chao Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | - Wanxiong He
- Inner Mongolia Medical University, Hohhot, China
| | - Leipeng Wang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | - Ting Hao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | - Xiaolong Yang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | - Wei Feng
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | | | - Chenyang Meng
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | - Zhi Wang
- Bayannur hospital, Bayannur, China
| | - Xiaofeng Chen
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Medical University, Hohhot, China
| | - Mingqi Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
- Inner Mongolia Medical University, Hohhot, China.
| | - Feng Zheng
- Department of Hepatic Hydatidosis, Qinghai Provincial People's Hospital, Xining, Qinghai, China.
| | - Baoxin Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
- Inner Mongolia Medical University, Hohhot, China.
- Tianjin Hospital, Tianjin University, Hexi District, Tianjin, China.
| |
Collapse
|
9
|
Shukla M, Narayan M. Proteostasis and Its Role in Disease Development. Cell Biochem Biophys 2025; 83:1725-1741. [PMID: 39422790 PMCID: PMC12123047 DOI: 10.1007/s12013-024-01581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Proteostasis (protein homeostasis) refers to the general biological process that maintains the proper balance between the synthesis of proteins, their folding, trafficking, and degradation. It ensures proteins are functional, locally distributed, and appropriately folded inside cells. Genetic information enclosed in mRNA is translated into proteins. To ensure newly synthesized proteins take on the exact three-dimensional conformation, molecular chaperones assist in proper folding. Misfolded proteins can be refolded or targeted for elimination to stop aggregation. Cells utilize different degradation pathways, for instance, the ubiquitin-proteasome system, the autophagy-lysosome pathway, and the unfolded protein response, to degrade unwanted or damaged proteins. Quality control systems of the cell monitor the folding of proteins. These checkpoint mechanisms are aimed at degrading or refolding misfolded or damaged proteins. Under stress response pathways, such as heat shock response and unfolded protein response, which are triggered under conditions that perturb proteostasis, the capacity for folding is increased, and degradation pathways are activated to help cells handle stressful conditions. The deregulation of proteostasis is implicated in a variety of illnesses, comprising cancer, metabolic diseases, cardiovascular diseases, and neurological disorders. Therapeutic strategies with a deeper insight into the mechanism of proteostasis are crucial for the treatment of illnesses linked with proteostasis and to support cellular health. Thus, proteostasis is required not only for the maintenance of cellular homeostasis and function but also for proper protein function and prevention of injurious protein aggregation. In this review, we have covered the concept of proteostasis, its mechanism, and how disruptions to it can result in a number of disorders.
Collapse
Affiliation(s)
- Manisha Shukla
- Department of Biotechnology, Pandit S.N. Shukla University, Shahdol, Madhya Pradesh, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
10
|
Yang Y, Li Q, Chu LT, Lin X, Chen H, Chen L, Tang J, Zeng T. Autophagy in cholangiocarcinoma: a comprehensive review about roles and regulatory mechanisms. Clin Transl Oncol 2025; 27:2391-2400. [PMID: 39585591 DOI: 10.1007/s12094-024-03797-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024]
Abstract
The role of autophagy in cholangiocarcinogenesis and its development is intricate. Autophagy has a dual role in cholangiocarcinoma, and understanding the function and mechanism of autophagy in cholangiocarcinoma is pivotal in guiding therapeutic approaches to its treatment in clinical settings. Recent studies have revealed that autophagy is involved in the complex biological behavior of cholangiocarcinoma. In this review, we have summarized the genes and drugs that would promote or inhibit autophagy, leading to change in cellular behaviors of cholangiocarcinoma, including apoptosis, proliferation, invasion and migration, and influence its cellular drug resistance. In addition, we concluded the signaling pathways modulating autophagy in cholangiocarcinoma cells, including PI3K/AKT/mTOR,p38MAPK,AMPK/mTOR,LKB1-AMPK, and AKT/WNK1, and ERK signaling pathways, which subsequently impacting apoptosis, death, migration, invasion, and proliferation. In conclusion, we would like that we can provide ideas for future cholangiocarcinoma treatment by comprehensively summarizing the latest studies on the relationship between autophagy and cholangiocarcinoma, including the factors affecting autophagy and related signaling pathways.
Collapse
Affiliation(s)
- Yuxia Yang
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Qiuyan Li
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Lok Ting Chu
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Xiaocong Lin
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Helian Chen
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Linsong Chen
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Jinjing Tang
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Renmin Rd, Xiashan District, Zhanjiang, Guangdong, 524000, People's Republic of China.
| |
Collapse
|
11
|
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
|
12
|
Wei M, Wu Y, Yang Q, Zhou Z, Xu X. Serum Starvation Regulates Autophagy of Human Periodontal Ligament Cells Through Reactive Oxygen Species Mediated Adenosine Monophosphate-Activated Protein Kinase/Mechanistic Target of RAPAMYCIN Axis. Int Dent J 2025; 75:1461-1471. [PMID: 40120460 PMCID: PMC11982979 DOI: 10.1016/j.identj.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/03/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
INTRODUCTION AND AIMS Human periodontal ligament cells (hPDLCs) play a pivotal role in periodontal tissue remodelling, a process essential for orthodontic tooth movement (OTM). Autophagy, a survival mechanism under cellular stress, is induced by nutrient deprivation and impacts hPDLC function. This study aimed to explore the role of autophagy in the adaptive response of hPDLCs to nutritional stress, an environment simulating conditions during OTM. METHODS Nutrient deprivation in hPDLCs was modelled through serum starvation. Autophagy levels and relevant markers were assessed using electron microscopy, protein assays, and gene expression analyses. Emphasis was placed on adenosine monophosphate-activated protein kinase (AMPK) signalling, specifically phosphorylation of AMPKα at Thr172, as a regulatory node in autophagy induction. Loss- and gain-of-function approaches were utilized to investigate the role of Thr172 in AMPK-mediated autophagy under nutrient stress. RESULTS Findings indicated a marked increase in reactive oxygen species-mediated autophagy in hPDLCs under nutrient deprivation. This process was significantly regulated by AMPK activation through Thr172 phosphorylation, establishing AMPK as a critical factor in autophagy induction during cellular adaptation to nutritional stress. CONCLUSION Nutritional stress enhances reactive oxygen species-mediated autophagy in hPDLCs via AMPK signalling, underscoring the role of autophagy in cellular adaptation during OTM. Targeting the AMPK pathway could provide novel insights for optimizing orthodontic treatment by leveraging cellular adaptive mechanisms. CLINICAL RELEVANCE Understanding the molecular mechanisms underlying autophagy in hPDLCs opens potential therapeutic pathways to improve OTM outcomes. Modulating autophagy may lead to advances in orthodontic therapies that facilitate periodontal tissue remodelling, enhancing clinical effectiveness.
Collapse
Affiliation(s)
- Mianxing Wei
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Yujie Wu
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Qian Yang
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Zheng Zhou
- University of Detroit Mercy, School of Dentistry, Graduate Periodontics, Detroit, Michigan, USA.
| | - Xiaomei Xu
- Department of Orthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, Sichuan, P.R. China.
| |
Collapse
|
13
|
Yan L, Quan Z, Sun T, Wang J. Autophagy signaling mediated by non-coding RNAs: Impact on breast cancer progression and treatment. Mol Aspects Med 2025; 103:101365. [PMID: 40305994 DOI: 10.1016/j.mam.2025.101365] [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: 11/26/2024] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025]
Abstract
Autophagy, a conserved cellular mechanism which detoxifies and degrades intracellular structures or biomolecules, has been identified as an important factor in the progression of human breast cancer and the development of treatment resistance. Non-coding RNAs (ncRNAs), a broad family of RNA, have the ability to influence various processes, including autophagy, due to their diverse downstream targets. ncRNAs play an important role in suppressing or activating autophagy by targeting autophagy-triggering components such as the ULK1 complex, Beclin1, and ATGs. Recent research has uncovered the intricate regulatory networks that govern autophagy dynamics, with ncRNAs emerging as key participants in this network. miRNAs, lncRNAs, and circRNAs are the three subfamilies of ncRNAs that have the most well-known interactions with autophagy, particularly macroautophagy. The high prevalence of breast cancer necessitates research into finding new biological processes that can help in early detection as well as enhance the effectiveness of treatment. The positive/negative link between autophagy and ncRNAs can be exploited as a supplementary therapy to improve sensitivity to treatment in breast cancer. This review investigates the regulatory roles of ncRNAs, particularly microRNAs (miRNAs), in modifying autophagy pathways in human breast cancer progression and treatment. However, future studies and clinical practice are needed to determine the most relevant microRNAs as biomarkers and also to better understand their role in breast cancer progression or treatment through modifying autophagy.
Collapse
Affiliation(s)
- Lei Yan
- Clinical Experimental Centre, Xi'an International Medical Center Hospital, No.777 Xitai Road, High-tech Zone, Xi'an, Shaanxi Province, 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptide, Xi'an, Shaanxi, 710100, China
| | - Zhuo Quan
- Clinical Experimental Centre, Xi'an International Medical Center Hospital, No.777 Xitai Road, High-tech Zone, Xi'an, Shaanxi Province, 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptide, Xi'an, Shaanxi, 710100, China
| | - Tiantian Sun
- Department of Oncology, Zibo Central Hospital, Shandong, 255036, China.
| | - Jiaju Wang
- Department of Hematology, Zibo Central Hospital, Shandong, 255036, China.
| |
Collapse
|
14
|
Suwannaphan T, Kamnerdsook A, Chalermwisutkul S, Techaumnat B, Damrongplasit N, Traipattanakul B, Kasetsirikul S, Pimpin A. Effects of Shear and Extensional Stresses on Cells: Investigation in a Spiral Microchannel and Contraction-Expansion Arrays. ACS Biomater Sci Eng 2025. [PMID: 40434022 DOI: 10.1021/acsbiomaterials.5c00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
In recent decades, inertial microfluidic devices have been widely used for cell separation. However, these techniques inevitably exert mechanical stresses, causing cell damage and death during the separation process. This remains a significant challenge for their biological and clinical applications. Despite extensive research on cell separation, the effects of mechanical stresses on cells in microfluidic separation have remained insufficiently explored. This review focuses on the effects of mechanical stresses on cells, particularly in spiral microchannels and contraction-expansion arrays (Contraction and Expansion Arrays (CEAs)). We derived the approximated magnitude of shear stress in a spiral microchannel, extensional stress in CEAs and conventional methods, along with exposure time in a single map to illustrate cell damage and operational zones. Finally, this review serves as a practical guideline to help readers in evaluating stress damages, enabling the effective selection of appropriate techniques that optimize cell viability and separation efficiency for biological and clinical applications.
Collapse
Affiliation(s)
- Thammawit Suwannaphan
- Department of Mechanical Engineering Technology, College of Industrial Technology (CIT), King Mongkut's University of Technology North Bangkok, Bangkok 10800, Thailand
- Center of Sustainable and Energy Engineering Materials, Department of Mechanical Engineering Technology, College of Industrial Technology, King Mongkut's University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Ampol Kamnerdsook
- Department of Mechanical Engineering Technology, College of Industrial Technology (CIT), King Mongkut's University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Suramate Chalermwisutkul
- The Sirindhorn International Thai-German Graduate School of Engineering (TGGS), King Mongkut's University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Boonchai Techaumnat
- Department of Electrical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
- Micro/Nano Electromechanical Integrated Device Research Unit, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nattapol Damrongplasit
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
- Micro/Nano Electromechanical Integrated Device Research Unit, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| | - Bhawat Traipattanakul
- School of Manufacturing Systems and Mechanical Engineering, Sirindhorn International Institute of Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Surasak Kasetsirikul
- Faculty of Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Alongkorn Pimpin
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
- Micro/Nano Electromechanical Integrated Device Research Unit, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
15
|
Zhang J, Wang W, Zhang X, Wang F, Geng S, Wang X, Wang T. Using endoplasmic reticulum engineering to improve recombinant protein production in CHO cells. Int J Biol Macromol 2025; 315:144695. [PMID: 40424905 DOI: 10.1016/j.ijbiomac.2025.144695] [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: 12/08/2024] [Revised: 05/22/2025] [Accepted: 05/25/2025] [Indexed: 05/29/2025]
Abstract
Chinese hamster ovary (CHO) cells are commonly used to produce recombinant therapeutic proteins (RTPs). While recent strategies have significantly improved the expression levels of RTPs in CHO cells, insufficient secretion and endoplasmic reticulum (ER) stress remain major bottlenecks. Therefore, further understanding of the mechanism of the ER stress response, optimization of ER-related folding and degradation pathways, and development of more efficient ER engineering tools are expected to overcome this issue and maximize RTP production. In this review, we summarize the role of ER in recombinant proteins production and explore ER engineering strategies to improve the yield of recombinant proteins in CHO cells. We further discuss ER-related strategies that can improve recombinant protein production, future research directions, and prospective applications.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China.
| | - Weifeng Wang
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Xi Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Fang Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Shaolei Geng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Xiaoyin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Tianyun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China.
| |
Collapse
|
16
|
Ho H, Aoki N, Eguchi M, Gao M, Suzuki H, Mikami M, Chiba H, Hui SP. Vitamin K2 Attenuates Mitochondrial Damage in Renal Proximal Tubular Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40400071 DOI: 10.1021/acs.jafc.5c01320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Excessive reactive oxygen species (ROS) levels cause oxidative stress, which can lead to various diseases. Renal failure is associated with oxidative stress and mitochondrial dysfunction. Vitamin K1 (phylloquinone) and K2 (menaquinone) are essential for blood coagulation and bone formation. Vitamin K has been shown to have anti-inflammation, glucose metabolism regulation, and antiferroptosis functions. We investigated the impact of menaquinone-4 (MK-4) on oxidative stress and mitochondrial dysfunction in human renal proximal tubular cells. MK-4 protected cells from oxidative damage induced by l-buthionine-(S,R)-sulfoximine (BSO), a selective inhibitor of glutathione metabolism, by inhibiting cell death, mitochondrial ROS production, and lipid peroxidation. MK-4 also reduced lactate production, prevented mitochondrial fragmentation, and improved mitochondrial respiratory function, indicating cytoprotective effects. Moreover, it enhanced intracellular ATP production and respiratory capacity, even in the absence of oxidative stress. Thus, MK-4 plays an important role in mitochondrial function in renal proximal tubular cells.
Collapse
Affiliation(s)
- Hsinjung Ho
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Natsumi Aoki
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Miki Eguchi
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Mingchen Gao
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Hirotaka Suzuki
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Mao Mikami
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| | - Hitoshi Chiba
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
- Department of Nutrition, Sapporo University of Health Sciences, Nakanuma Nishi-4-2-1-15, Higashi-ku, Sapporo 007-0894, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
17
|
Meng X, Gong Y, Xiao F, Cao Z, Zhuang Z, Yi X, Wang J, Feng R, Gong C, Ni P. Curcumin's multi-target mechanisms in the treatment of Alzheimer's disease and creative modification techniques. J Alzheimers Dis 2025:13872877251344188. [PMID: 40397414 DOI: 10.1177/13872877251344188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Alzheimer's disease (AD) is a well-established neurodegenerative disorder characterized by memory impairment, cognitive dysfunction, and behavioral disturbances. With the global population aging, the prevalence of AD continues to rise, presenting significant challenges to both society and healthcare systems. Curcumin, a polyphenolic compound derived from turmeric rhizomes, has demonstrated considerable potential in AD treatment due to its anti-inflammatory, antioxidant, and neuroprotective properties. However, its clinical application remains constrained by chemical instability, poor water solubility, rapid metabolism, and accelerated elimination. To overcome these limitations, various curcumin derivatives have been synthesized, and combination therapy strategies have been explored. This review examines the potential mechanisms through which curcumin may exert therapeutic effects in AD, including the inhibition of neuroinflammation, regulation of tau protein hyperphosphorylation, modulation of amyloid-β peptides, and provision of antioxidant benefits. Additionally, the advantages of curcumin derivatives and combination therapy approaches are discussed, offering novel perspectives and promising strategies for AD treatment. It is anticipated that advancements in drug design and therapeutic approaches will contribute to the development of more effective treatment options for AD.
Collapse
Affiliation(s)
- Xiaoyuan Meng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Yong Gong
- Hainan General Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Fengxin Xiao
- Hainan General Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Zhao Cao
- Hainan General Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Zheyu Zhuang
- Hainan General Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Xinan Yi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Juan Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Renjun Feng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Chunmei Gong
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| | - Panli Ni
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
18
|
Liu X, Liu X, Zhao L, Wu J, Wang X, Hu Y. RIP2/NF-κB/PD-L1 signaling pathway is involved in temozolomide resistance by inducing autophagy in glioblastoma cells. Transl Oncol 2025; 58:102424. [PMID: 40403475 DOI: 10.1016/j.tranon.2025.102424] [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: 12/15/2024] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025] Open
Abstract
Autophagy is an important factor in temozolomide (TMZ) resistance in glioblastoma (GBM). Receptor-interacting protein 2 (RIP2) is associated with autophagy, but its role and mechanism in regulating autophagy in GBM cells remain unclear. To analyze RIP2 expression in GBM in The Cancer Genome Atlas (TCGA) dataset. GBM cells were stimulated using recombinant human RIP2 protein (rRIP2) or RIP2 plasmid. Cell proliferation and apoptosis were assessed using CCK-8 assay and flow cytometry. Western blotting and immunofluorescence (IF) assays were performed to detect protein expression in cells and tumor tissues. Moreover, the relationship between RIP2-induced autophagy and TMZ resistance was verified in a GBM xenograft model. We determined that RIP2 expression was upregulated in GBM. rRIP2 and RIP2 overexpression induced TMZ resistance in the GBM cell lines. RIP2 overexpressing xenograft tumors have reduced sensitivity to TMZ. In addition, we showed that PD-L1 protein expression was upregulated in GBM tissues with RIP2 overexpression. rRIP2 and RIP2 overexpression induced autophagy in GBM cells through AMPK. Notably, RIP2 upregulated PD-L1 expression through the NF-κB signaling pathway, which induced autophagy and TMZ resistance in GBM cells. Moreover, NF-κB or autophagy inhibition reversed TMZ resistance in RIP2 overexpressing GBM cells in a xenograft model. In conclusion, RIP2 induces TMZ resistance in GBM cells by promoting autophagy through the NF-κB/PD-L1 signaling pathway, indicating that the RIP2/NF-κB/PD-L1 pathway may be therapeutic target for TMZ resistance.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Xiaosong Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Lei Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jianliang Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Xiaoliang Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Yuhua Hu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China.
| |
Collapse
|
19
|
Antonakoudis A, Kyriakoudi SA, Chatzi D, Dermitzakis I, Gargani S, Meditskou S, Manthou ME, Theotokis P. Genetic Basis of Motor Neuron Diseases: Insights, Clinical Management, and Future Directions. Int J Mol Sci 2025; 26:4904. [PMID: 40430041 PMCID: PMC12112488 DOI: 10.3390/ijms26104904] [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: 03/27/2025] [Revised: 05/03/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Motor neuron diseases (MNDs) are a heterogeneous group of neurodegenerative disorders characterized by the progressive loss of motor neurons, resulting in debilitating physical decline. Advances in genetics have revolutionized the understanding of MNDs, elucidating critical genes such as SOD1, TARDBP, FUS, and C9orf72, which are implicated in their pathogenesis. Despite these breakthroughs, significant gaps persist in understanding the interplay between genetic and environmental factors, the role of rare variants, and epigenetic contributions. This review synthesizes current knowledge on the genetic landscape of MNDs, highlights challenges in linking genotype to phenotype, and discusses the promise of precision medicine approaches. Emphasis is placed on emerging strategies, such as gene therapy and targeted molecular interventions, offering hope for personalized treatments. Addressing these challenges is imperative to harness the full potential of genomics for improving outcomes in MNDs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.A.); (S.A.K.); (D.C.); (I.D.); (S.G.); (S.M.); (M.E.M.)
| |
Collapse
|
20
|
Wang H, Feng X, He H, Li L, Wen Y, Liu X, He B, Hua S, Sun S. Crosstalk between autophagy and other forms of programmed cell death. Eur J Pharmacol 2025; 995:177414. [PMID: 39986593 DOI: 10.1016/j.ejphar.2025.177414] [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: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Cell death occurs continuously throughout individual development. By removing damaged or senescent cells, cell death not only facilitates morphogenesis during the developmental process, but also contributes to maintaining homeostasis after birth. In addition, cell death reduces the spread of pathogens by eliminating infected cells. Cell death is categorized into two main forms: necrosis and programmed cell death. Programmed cell death encompasses several types, including autophagy, pyroptosis, apoptosis, necroptosis, ferroptosis, and PANoptosis. Autophagy, a mechanism of cell death that maintains cellular equilibrium via the breakdown and reutilization of proteins and organelles, is implicated in regulating almost all forms of cell death in pathological contexts. Notably, necroptosis, ferroptosis, and PANoptosis are directly classified as autophagy-mediated cell death. Therefore, regulating autophagy presents a therapeutic approach for treating diseases such as inflammation and tumors that arise from abnormalities in other forms of programmed cell death. This review focuses on the crosstalk between autophagy and other programmed cell death modalities, providing new perspectives for clinical interventions in inflammatory and neoplastic diseases.
Collapse
Affiliation(s)
- Huaiyuan Wang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China; Clinical Medicine, class 3, 2022 Grade, Kunming Medical University, Kunming, China
| | - Xiran Feng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China; Clinical Medicine, Kunming Medical University-Shanghai Jiaotong University Joint Program, 2022 Grade, Kunming Medical University, Kunming, China
| | - Huilin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Lingyu Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaofei Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Bifeng He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China.
| |
Collapse
|
21
|
Wolska W, Gutowska I, Wszołek A, Żwierełło W. The Role of Intermittent Fasting in the Activation of Autophagy Processes in the Context of Cancer Diseases. Int J Mol Sci 2025; 26:4742. [PMID: 40429883 PMCID: PMC12112746 DOI: 10.3390/ijms26104742] [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: 04/27/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Intermittent fasting (IF) is a dietary approach that influences key metabolic pathways, including autophagy-a crucial mechanism in maintaining cellular homeostasis. Autophagy plays a dual role in oncogenesis, acting both as a tumor suppressor and a survival mechanism under metabolic stress. IF has shown potential for reducing cancer risk and enhancing therapeutic efficacy by sensitizing tumor cells to chemotherapy and radiotherapy. However, its effects depend heavily on the type and stage of cancer. Potential risks, such as excessive weight loss and malnutrition, require careful evaluation. Further clinical studies are needed to optimize IF protocols as adjuncts to cancer therapy. This review discusses autophagy mechanisms induced by IF, their therapeutic implications in oncology, and the limitations of this dietary strategy.
Collapse
Affiliation(s)
- Waleria Wolska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (W.W.); (W.Ż.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030 Trondheim, Norway
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (W.W.); (W.Ż.)
| | - Agata Wszołek
- Institute of Biology, University of Szczecin, Wąska 13, 71-415 Szczecin, Poland;
| | - Wojciech Żwierełło
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (W.W.); (W.Ż.)
| |
Collapse
|
22
|
Liu C, Wang X, Xu S, Liu M, Cao X. Regulation of autophagy: Insights into O-GlcNAc modification mechanisms. Life Sci 2025; 369:123547. [PMID: 40058573 DOI: 10.1016/j.lfs.2025.123547] [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: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/30/2025]
Abstract
Autophagy is a "self-eating" biological process that degrades cytoplasmic contents to ensure cellular homeostasis. Its response to stimuli occurs in two stages: Within a few to several hours of exposure to a stress condition, autophagic flow rapidly increases, which is mediated by post-translational modification (PTM). Subsequently, the transcriptional program is activated and mediates the persistent autophagic response. O-linked β-N-acetylglucosamine (O-GlcNAc) modification is an inducible and dynamically cycling PTM; mounting evidence suggests that O-GlcNAc modification participates in the total autophagic process, including autophagy initiation, autophagosome formation, autophagosome-lysosome fusion, and transcriptional process. In this review, we summarize the current knowledge on the emerging role of O-GlcNAc modification in regulating autophagy-associated proteins and explain the different regulatory effects on autophagy exerted by O-GlcNAc modification.
Collapse
Affiliation(s)
- Chengzhi Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinyu Wang
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Shengnan Xu
- College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Mingyue Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xusheng Cao
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
23
|
Malik JA, Agrewala JN. Assessing the Implications of Morphine-Induced Dysregulation of Autophagy on Brain Health. Mol Neurobiol 2025:10.1007/s12035-025-05039-5. [PMID: 40355798 DOI: 10.1007/s12035-025-05039-5] [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: 10/30/2024] [Accepted: 05/04/2025] [Indexed: 05/15/2025]
Abstract
Morphine has been a widely used drug for pain management and anesthesia in clinical settings for centuries and is also a drug of abuse. Its illicit use by individuals with substance use disorders has resulted in numerous brain-related complications. The immunopharmacology of morphine is highly complex, necessitating a deeper understanding of its interactions with brain regions involved in learning and memory. Autophagy is a conserved physiological recycling process that degrades cytoplasmic organelles and proteins, repurposing their components for cellular function. However, recent studies indicate that morphine exposure disrupts autophagic processes, contributing to many morphine-associated complications. This article highlights recent advancements in understanding the interplay between morphine and autophagy. By exploring this intricate relationship, we aim to enhance our knowledge of morphine-associated complications and autophagy dysregulation, potentially improving the management of morphine use disorder and related conditions, thereby promoting healthier outcomes.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India
| | - Javed N Agrewala
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, 140001, India.
| |
Collapse
|
24
|
Wang F, Lei Z, Gao J, Li Z, Liu S, Wan J, Lei Z. P-glycoprotein inhibits the MAPK/NF-KB pathway and activates autophagy and oxidative stress to improve GPS resistance in vivo and in vitro. Int J Biol Macromol 2025; 314:144126. [PMID: 40368213 DOI: 10.1016/j.ijbiomac.2025.144126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/26/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Glaesserella parasuis (Gps) is an important pathogen that causes polyserositis, peritonitis and meningitis in pigs, causing considerable economic losses to the pig industry worldwide. In recent years, due to the abuse of antibiotic drugs, Gps drug resistance and multi-drug resistance have become increasingly serious, especially for macrolides. Thus, by inducing Tydigrosin resistant bacteria, we found that PgtP is a Gps-produced virulence gene that plays a crucial role in the disease, but its interaction in host cells remains unclear. To characterize the function of the PgtP gene in Gps, we constructed ΔPgtP mutants. Notably, deletion of the PgtP gene resulted in increased adhesion to mouse macrophages, suggesting that PgtP is essential for adhesion of Gps. In addition, as a member of the cell membrane transporter family, PgtP links activated inflammation with autophagy and oxidative stress metabolism. To further investigate the role of PgtP, we infected mouse macrophages with ΔPgtP mutants and wild strains respectively. We found that ΔPgtP mutations can reduce the level of intracellular reactive oxygen species (ROS), up-regulate the number of intracellular autophagosomes, and down-regulate the expression of cytokines IL-6 and TNF-a. Further mechanistic studies showed that PGTP-associated p38 mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways were involved in autophagy and oxidative stress and demonstrated that the related signaling pathways were also activated in mice. These results highlight the potential of PgtP as a target for therapeutic intervention and provide important new insights into how it contributes to the aggressiveness and persistence of Gps.
Collapse
Affiliation(s)
- Fan Wang
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhiqun Lei
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingyi Gao
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Ze Li
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Shenglan Liu
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Juan Wan
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Gannan Innovation and Translational Medicine Research Institute, First Affiliated Hospital, Gannan Medical University, Ganzhou 341000, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Sichuan Clinical Research Center for Medical Imaging, Dazhou, 635000, China; Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
25
|
Wang J, Shan J, Guo C, Duan Y, Zhang F, Ye W, Liu Y. Transcriptome analysis and machine learning methods reveal potential mechanisms of zebrafish muscle aging. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101532. [PMID: 40367591 DOI: 10.1016/j.cbd.2025.101532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
Muscle is one of the most abundant tissues in the human body, and its aging usually leads to many adverse consequences. Zebrafish is a powerful model used to study human muscle diseases, yet we know little about the molecular mechanisms of muscle aging in zebrafish. In this study, we determined the gene expression profiles of muscle tissues from male zebrafish of four different ages. Through differential expression analysis and expression pattern analysis, we identified a set of genes associated with muscle aging in zebrafish. Functional enrichment analysis revealed that several biological changes accompanied zebrafish muscle aging, including chronic inflammation, accumulation of sphingolipids, reduction of autophagy, and activation of the ferroptosis pathway. H&E staining showed that zebrafish muscle senescence leads to myofibrillar interstitial expansion and inflammatory cell infiltration. Furthermore, we screened zebrafish muscle aging related biomarkers by machine learning and verified the expression levels of some biomarkers by RT-qPCR. Based on these biomarkers, we constructed a zebrafish muscle aging clock that can predict muscle age based on transcriptomic data. This study provides us with a new perspective to understand the molecular mechanism of muscle aging and a new tool for zebrafish-based anti-aging research.
Collapse
Affiliation(s)
- Jian Wang
- Anesthesiology Department, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, Zhejiang, China
| | - Junwei Shan
- Hunter Biotechnology, Inc., Hangzhou 310051, China
| | - Cheng Guo
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - You Duan
- West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Zhang
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| | - Weidong Ye
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| | - Yanxiao Liu
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| |
Collapse
|
26
|
Shang F, Nie H, Du L, Shang J, Song X, Chen Y, Li H, Wang Z, Qi Y, Zhao L. Inhibition of ATG5-mediated autophagy maintains PMAIP1 stability to promote cell apoptosis and suppress triple-negative breast cancer progression. Discov Oncol 2025; 16:687. [PMID: 40338396 PMCID: PMC12061822 DOI: 10.1007/s12672-025-02470-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer characterized by a high recurrence rate and a lack of effective targeted therapies. The purpose of this study was to investigate the interaction between the pro-apoptotic factor phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) and autophagy-related protein 5 (ATG5), as well as their regulatory mechanisms in TNBC cell apoptosis and autophagy, to identify potential therapeutic targets for TNBC. METHODS TNBC-related datasets were retrieved from The Cancer Genome Atlas and selected by Prediction Analysis of Microarray 50 analysis to assess the expression of PMAIP1 in the samples. Additionally, the expression of PMAIP1 in the TNBC cell lines (MDA-MB-231) was detected using quantitative real-time polymerase chain reaction. In MDA-MB-231 cells, the expression of PMAIP1 and ATG5 was overexpressed or knocked down, and autophagy was inhibited using chloroquine (20 μM). Gene and protein expression levels were evaluated using quantitative real-time polymerase chain reaction and Western blot, respectively. Immunofluorescence was used to observe microtubule-associated protein 1 light chain 3 puncta formation to assess autophagy levels. Furthermore, cell apoptosis, proliferation, migration, and invasion were analyzed using terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, colony formation assay, and Transwell assay. RESULTS Compared to the control group, the expression of PMAIP1 was significantly elevated in TNBC tissues and MDA-MB-231 cells. Furthermore, overexpression of PMAIP1 led to a marked increase in apoptosis levels and a remarkable reduction in autophagy levels in MDA-MB-231 cells, while knockdown of PMAIP1 showed the opposite effects. Additionally, knockdown of ATG5 expression or treatment with chloroquine not only resulted in an increase in PMAIP1 expression in a time-dependent manner, but also reduced autophagy levels and enhanced apoptosis levels of cells. Furthermore, simultaneous knockdown of PMAIP1 and ATG5 considerably up-regulated apoptosis levels while down-regulating autophagy levels. Moreover, knockdown of PMAIP1 alone promoted the viability, invasion, and migration abilities of TNBC cells, while dual knockdown reversed these effects. CONCLUSION Inhibition of ATG5-mediated autophagy maintains PMAIP1 stability, thereby promoting cell apoptosis and suppressing TNBC progression.
Collapse
Affiliation(s)
- Fangjian Shang
- Department of General Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, 050023, Hebei, China
- Department of Gland Surgery, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Hongfeng Nie
- Department of Gland Surgery, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Liyan Du
- Department of Gland Surgery, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Jin Shang
- Department of Gland Surgery, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China
| | - Xiangquan Song
- School of Basic Medical Sciences, Xingtai Medical College, Xingtai, 054000, Hebei, China
| | - Ya Chen
- School of Basic Medical Sciences, Xingtai Medical College, Xingtai, 054000, Hebei, China
| | - Hui Li
- School of Basic Medical Sciences, Xingtai Medical College, Xingtai, 054000, Hebei, China
| | - Zhuo Wang
- School of Basic Medical Sciences, Xingtai Medical College, Xingtai, 054000, Hebei, China
| | - Yixin Qi
- Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
| | - Liyan Zhao
- Department of Gland Surgery, Hebei General Hospital, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
27
|
Pu N, Li S, Wu H, Zhao N, Wang K, Wei D, Wang J, Sha L, Zhao Y, Tao Y, Song Z. Beacon of Hope for Age-Related Retinopathy: Antioxidative Mechanisms and Pre-Clinical Trials of Quercetin Therapy. Antioxidants (Basel) 2025; 14:561. [PMID: 40427443 PMCID: PMC12108410 DOI: 10.3390/antiox14050561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Age-related retinopathy is one of the leading causes of visual impairment and irreversible blindness, characterized by progressive neuronal and myelin loss. The damages caused by oxidation contributes to the hallmarks of aging and represents fundamental components in pathological pathways that are thought to drive multiple age-related retinopathies. Quercetin (Que), a natural polyphenol abundant in vegetables, herbs, and fruits, has been extensively studied for its long-term antioxidative effects mediated through diverse mechanisms. Additionally, Que and its derivatives exhibit a broad spectrum of pharmacological characteristics in the cellular responses of age-related retinopathy induced by oxidative stress, including anti-inflammatory, anti-neovascularization, regulatory, and neuroprotective effects in autophagy and apoptosis processes. This review mainly focuses on the antioxidative mechanisms and curative effects of Que treatment for various age-related retinopathies, such as retinitis pigmentosa, diabetic retinopathy, age-related macular degeneration, and glaucoma. Furthermore, we discuss emerging technologies and methods involving Que and its derivatives in the therapeutic strategies for age-related retinopathies, highlighting their promise for clinical translation.
Collapse
Affiliation(s)
- Ning Pu
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Hao Wu
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Na Zhao
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Kexin Wang
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Dong Wei
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Jiale Wang
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Lulu Sha
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Yameng Zhao
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Ye Tao
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (N.Z.); (K.W.)
| | - Zongming Song
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, China; (N.P.); (H.W.); (D.W.); (J.W.); (L.S.); (Y.Z.)
| |
Collapse
|
28
|
Lee J, Park JR, Lee H, Hong SH, Kim WJ, Eickelberg O, Park SM, Ryu S, Cho SJ, Kim SJ, Yang SR. Fludarabine attenuates inflammation and dysregulated autophagy in alveolar macrophages via inhibition of STAT1/IRF1 pathway. Lab Anim Res 2025; 41:12. [PMID: 40336064 PMCID: PMC12057031 DOI: 10.1186/s42826-025-00245-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/09/2025] [Accepted: 04/27/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Acute lung injury (ALI), including its most severe form, acute respiratory distress syndrome (ARDS), is a common cause of acute hypoxemic respiratory failure. Although its clinical characteristics have been well characterized, the relevant mechanism remains unclear. An imbalance in autophagy leads to alveolar remodeling and triggers the pathogenesis of ARDS. In this study, we assessed the therapeutic efficacy of the STAT1 inhibitor fludarabine (Fluda) in ALI. C57BL6 mice were exposed to lipopolysaccharide (LPS), and their lung tissues were analyzed via next-generation transcriptome sequencing. RESULTS Western blotting revealed that interferon regulatory factor 1 (IRF1) was highly expressed and STAT1 was phosphorylated following LPS exposure. Fluda significantly decreased the protein expression of STAT1/IRF1 and inhibited the alveolar infiltration of neutrophils and macrophages. Nitric oxide (NO), inducible nitric oxide synthase, tumor necrosis factor-α (TNF-α), interferon-γ, and interleukin-6 (IL-6) release was decreased in the lungs of mice and RAW264.7 macrophages following Fluda treatment. In LPS-induced GFP-LC3 transgenic mice treated with Fluda, the counts of LC3-expressing neutrophils and macrophages in bronchoalveolar (BAL) fluid were significantly decreased. Furthermore, Fluda decreased LC3 and p62 protein expression, thereby inhibiting the release of NO, IL-6, and TNF-α in BAL. In RAW264.7 cells, the inhibition of STAT1/IRF1 by Fluda decreased LPS-induced ERK and NF-κB p65 phosphorylation. CONCLUSIONS The inhibition of STAT1/IRF1 by Fluda plays a pivotal role in modulating dysregulated autophagy by suppressing the MAPK and NF-κB p65 pathways in ALI.
Collapse
Affiliation(s)
- Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Jeong-Ran Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Hanbyeol Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Oliver Eickelberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sung-Min Park
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Semin Ryu
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Sung Joon Cho
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341, Chuncheon, South Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, Gangwon, South Korea.
- Institute of Medical Science, School of Medicine, Kangwon National University, Gangwon State, 1 Kanwondaehak-Gil, Chuncheon, 24341, South Korea.
| |
Collapse
|
29
|
Tian S, Xu H, Wu X, Ding Y, Liang L, Yin H, Zeng X, Liu Y, Zhu W. Ruthenium(II) polypyridyl complexes inhibit tumor growth through stimulating immune system to increase CD8 + T cell. Eur J Med Chem 2025; 289:117470. [PMID: 40054298 DOI: 10.1016/j.ejmech.2025.117470] [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: 12/23/2024] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025]
Abstract
In this work, we have carefully designed and synthesized two Ru(II) metal complexes: [Ru(phen)2(HMPIP)](PF6)2 (6a, where phen = 1,10-phenanthroline, HMPIP = 2-(2-hydroxy-3-methylphenyl-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ru(bpy)2(HMPIP)](PF6)2 (6b, where bpy = 2,2'-bipyridine). Using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to explore the cytotoxicity of 6a and 6b towards HepG2, B16, A549, SGC-7901, HCT116 and non-cancer LO2. The complexes exhibited cytotoxicity activity against HepG2 cells. The capacity of 6a and 6b to impede the proliferation and dissemination of cancer cells was evaluated by conducting proliferation and migration experiments and 3D model. The anticancer mechanism was investigated in detail. The utilization of cycle blocking assays revealed that 6a and 6b induced a G0/G1 phase arrest in HepG2 cells. The cellular uptake experiments show that the complexes enter the cell nuclei, then escape from the cell nuclei into the cytoplasm, finally accumulate in the mitochondria. Apoptosis assays and the examination of proteins indicated that the complexes were capable of efficiently inducing apoptosis in HepG2 cells. Additionally, the potential induction of autophagy-mediated cell death was explored. The observed reduction in glutathione (GSH) levels and glutathione peroxidase 4 (GPX4) expression suggested a disruption of redox homeostasis within cancer cells, an increment in malondialdehyde (MDA) amount, together with BODIPY staining experiment, confirm that 6a and 6b can induce ferroptosis. Interestingly, in a nude mouse model, 6a showed a significant suppression of tumor growth with an inhibition rate of 63.4 %, without causing any weight loss of mice. The studies on the mechanism show that 6a causes immune cell death, increase the amount of TNF-α and IFN-γ, reduce IL-10 content, which further activates immune response to increase CD8+ T cells to prevent tumor growth. Therefore, 6a inhibits the tumor growth through stimulating the immune response to increase CD8+ T cells. In addition, the experiments in vitro show that the complexes through inhibition of PI3K/AKT/mTOR signaling pathway and intrinsic mitochondria pathway to cause cell apoptosis. These results demonstrate that Ru(II) complexes may be potent anticancer candidates for HepG2 tumor.
Collapse
Affiliation(s)
- Shuang Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Haixin Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Xiaoyu Wu
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yueyao Ding
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Xiandong Zeng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Wenrun Zhu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
30
|
Li Y, Gao A, Zhou W, Tang X, Zeng T, Fan T, Jiang W, Tang M, Ouyang F. Unveiling the protective role of ESM1 in endothelial cell proliferation and lipid reprogramming. Sci Rep 2025; 15:15572. [PMID: 40320451 PMCID: PMC12050336 DOI: 10.1038/s41598-025-00581-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Palmitic acid (PA), being the most prevalent free fatty acid in the human, holds significant implications as a risk factor for atherosclerosis (AS) due to its ability to induce physiological dysfunction in endothelial cells (ECs). Endothelial cell-specific molecule 1 (ESM1), has been identified as a marker for activated ECs. Nevertheless, the mechanisms underlying ESM1-induced endothelial cell proliferation remain elusive. The expression of ESM1, ANGPTL4 and autophagy related protein were confirmed by western blot. Proliferation ability was tested by MTT and EdU. Lipids level was confirmed by Oil red staining. Autophagic flux was confirmed by Monodansylcadaverine (MDC) staining and pCMV-mCherry-GFP-LC3B fluorescence staining assay. The mouse model of AS was used to observe the effect of PA on the ESM1-ANGPTL4-autophagy signaling axis. This study elucidates ESM1-ANGPTL4 axis in maintaining proliferation of ECs and lipid reprogramming. Furthermore, it has been observed that PA has the ability to stimulate EC to autonomously increase the expression of ESM1, which in turn can counteract the detrimental effects of PA on ECs. Conversely, when ESM1 is suppressed, the damaging effects of PA on ECs are exacerbated. Mechanistically, our findings indicate that ESM1 facilitates EC proliferation and lipids homeostasis by up-regulating autophagy through ANGPTL4. This effect of ESM1 on ECs can be attenuated by ATG7 inhibiting. Additionally, the serum levels of ESM1 were found to be elevated in AS mice. ESM1 was found to enhance ECs proliferation and mitigate endothelial cell injury induced by PA through the upregulation of autophagy. This mechanism potentially serves as a protective factor against atherosclerosis progression.
Collapse
Affiliation(s)
- Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Anbo Gao
- Department of Cardiology, Xiangya Hospital Zhuzhou, Zhuzhou Central Hospital, Central South University, Central South University, Zhuzhou, Hunan, China
- Clinical Research Institute, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenchao Zhou
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
| | - Tian Zeng
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tingyu Fan
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weimin Jiang
- Department of Cardiology, Xiangya Hospital Zhuzhou, Zhuzhou Central Hospital, Central South University, Central South University, Zhuzhou, Hunan, China
| | - Min Tang
- Department of Cardio-Thoracic Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fan Ouyang
- Department of Cardiology, Xiangya Hospital Zhuzhou, Zhuzhou Central Hospital, Central South University, Central South University, Zhuzhou, Hunan, China.
- Department of Cardiology, Zhuzhou Hospital, the Affiliated Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China.
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
31
|
Pan R, Koo C, Su W, You Q, Guo H, Liu B. Circular RNAs modulate cell death in cardiovascular diseases. Cell Death Discov 2025; 11:214. [PMID: 40316538 PMCID: PMC12048724 DOI: 10.1038/s41420-025-02504-x] [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: 02/06/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a global health challenge, with programmed cell death (PCD) mechanisms like apoptosis and necroptosis playing key roles in the progression. Circular RNAs (circRNAs) have recently been recognized as crucial regulators of gene expression, especially in modulating PCD. In current researches, circRNA regulation of apoptosis is the most studied area, followed by autophagy and ferroptosis. Notably, the regulatory role of circRNAs in pyroptosis and necroptosis has also begun to attract attention. From a mechanistic perspective, circRNAs influence cellular processes through several modes of action, including miRNA sponging, protein interactions, and polypeptide translation. Manipulating circRNAs and their downstream targets through inhibition or overexpression offers versatile therapeutic options for CVD treatment. Continued investigation into circRNA-mediated mechanisms may enhance our understanding of CVD pathophysiology and underscore their potential as novel and promising therapeutic targets.
Collapse
Affiliation(s)
- Runfang Pan
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chinying Koo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenyuan Su
- Sport Medicine & Rehabilitation Center, Shanghai University of Sport, Shanghai, 200438, China
| | - Qianhui You
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haidong Guo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
32
|
Kaur S, Vashistt J, Kumar A, Parkash J, Changotra H. Autophagy-related gene BECN1 single nucleotide polymorphisms in diseases. Mol Cell Biochem 2025; 480:2765-2769. [PMID: 39636430 DOI: 10.1007/s11010-024-05177-9] [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: 06/10/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Autophagy is a cytoprotective process that operates within a cell to maintain cellular homeostasis. An array of multiple proteins is involved to mediate this conserved cellular process. Among these, Beclin 1 protein encoded by BECN1 gene plays a crucial role during the initiation of autophagy. It acts as a molecular platform onto which multiple proteins interact to mediate autophagy initiation. The functioning of such proteins has reportedly been influenced by the molecular markers such as Single Nucleotide Polymorphisms (SNPs) present within the encoding gene. The SNPs within the autophagy gene have been known to influence the functioning of autophagy proteins which further is involved in various diseases. Studies have reported that the SNPs within the BECN1 are involved in various diseases. This report outlines the findings of all the existing research on the role of SNPs within BECN1.
Collapse
Affiliation(s)
- Sargeet Kaur
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Jitendraa Vashistt
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Ajay Kumar
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, 121001, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151 001, India
| | - Harish Changotra
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab, 143 005, India.
| |
Collapse
|
33
|
Liu M, Wang Y, Ren F, Zhang W, Zheng H, Zhang R, Gao C, Luo L, Nie C, Gu J. Simulated microgravity activates autophagy expression in the rat retina. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:107-116. [PMID: 40280632 DOI: 10.1016/j.lssr.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVE This study aims to investigate the expression and possible role of autophagy in the retina of rats under microgravity. METHODS Adult Sprague-Dawley (SD) rats were randomly allocated to either the tail suspension group (TS) or the control group (CTRL). To simulate microgravity-induced redistribution of cephalad fluid observed in space, the rats in the TS group underwent tail suspension for a duration of 4 weeks. Optical coherence tomography angiography (OCTA) was applied to assess the ocular blood flow and thickness of the retina. Hematoxylin and eosin (H&E) staining, along with transmission electron microscopy (TEM), were used to investigate morphological changes and autophagosomes in the retina. Endoplasmic reticulum autophagy (ER-phagy) related proteins (ATF4, CHOP, and GRP78) in the rat retina were detected using an immunofluorescence assay (IFA). The levels of autophagy-related proteins (Beclin1, P62, LC3B, ATF4, CHOP, and GRP78) were quantified by Western blot (WB). The expression of ATG5 and ATG7 genes was examined via real-time quantitative PCR (qPCR). RESULTS In fundus imaging signs, microgravity increases retinal thickness and the retinal vascular perfusion area. Moreover, microgravity also upregulates Beclin1, LC3B, ATF4, CHOP, and GRP78 while downregulating P62 in retina. It elevates the number of autophagosomes and activates autophagy and ER-phagy signaling pathways in retina. CONCLUSION Simulated microgravity can trigger the organism's intrinsic protective mechanisms, inducing the activation of autophagy (ER-phagy) in the retina, which may represent a self-defense mechanism against adverse conditions of microgravity-related stressors.
Collapse
Affiliation(s)
- Meng Liu
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuyu Wang
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Ren
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Wenqian Zhang
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Hanwen Zheng
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Rong Zhang
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Caiyun Gao
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Ling Luo
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China
| | - Chuang Nie
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China.
| | - Jianwen Gu
- The Ninth Medical Center of PLA General Hospital, Beijing, 100101, China.
| |
Collapse
|
34
|
Liao D, He Y, He B, Zeng S, Cui Y, Li C, Huang H. Inhibiting SNX10 induces autophagy to suppress invasion and EMT and inhibits the PI3K/AKT pathway in cervical cancer. Clin Transl Oncol 2025; 27:2084-2094. [PMID: 39367898 DOI: 10.1007/s12094-024-03715-x] [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: 06/02/2024] [Accepted: 09/02/2024] [Indexed: 10/07/2024]
Abstract
PURPOSE Cervical cancer (CC) is a prevalent malignancy among women with high morbidity and poor prognosis. Sorting nexin 10 (SNX10) is a newly recognized cancer regulatory factor, while its action on CC progression remains elusive. Hence, this study studied the effect of SNX10 on CC development and investigated the mechanism. METHODS The SNX10 level in CC and the overall survival of CC cases with different SNX10 expressions were determined by bioinformatics analysis in GEPIA. The SNX10 expression in tumor tissues and clinical significance were studied in 64 CC cases. The overall survival was assessed using Kaplan-Meier analysis. The formation of LC3 was evaluated using immunofluorescence. Cell invasion was measured using the Transwell assay. Epithelial-to-mesenchymal transition (EMT) was determined by observing cell morphology and assessing EMT marker levels. A xenograft tumor was constructed to evaluate tumor growth. RESULTS SNX10 was elevated in CC tissues and cells, and the CC cases with high SNX10 levels exhibited poor overall survival. Besides, SNX10 correlated with the FIGO stage, lymph node invasion, and stromal invasion of CC. SNX10 silencing induced CC cell autophagy and suppressed CC cell invasion and EMT. Meanwhile, silenced SNX10 could suppress invasion and EMT via inducing autophagy. Furthermore, SNX10 inhibition suppressed the PI3K/AKT pathway. Moreover, silenced SNX10 restrained the tumor growth, autophagy, and EMT of CC in vivo. CONCLUSION SNX10 was enhanced in CC and correlated with poor prognosis. Silenced SNX10 induced autophagy to suppress invasion and EMT and inhibited the PI3K/AKT pathway in CC, making SNX10 a valuable molecule for CC therapy.
Collapse
Affiliation(s)
- Dan Liao
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China.
| | - Yanxian He
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Bin He
- Clinical Translational Medical Center, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Saitian Zeng
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Yejia Cui
- Department of Clinical Laboratory, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Cuifen Li
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Haohai Huang
- Clinical Translational Medical Center, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China.
- Department of Clinical Pharmacy, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China.
| |
Collapse
|
35
|
S Y, K L M, Harithpriya K, Zong C, Sahabudeen S, Ichihara G, Ramkumar KM. Disruptive multiple cell death pathways of bisphenol-A. Toxicol Mech Methods 2025; 35:430-443. [PMID: 39815394 DOI: 10.1080/15376516.2024.2449423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
Endocrine-disrupting chemicals (EDCs) significantly contribute to health issues by interfering with hormonal functions. Bisphenol A (BPA), a prominent EDC, is extensively utilized as a monomer and plasticizer in producing polycarbonate plastic and epoxy resins, making it one of the highest-demanded chemicals in commercial use. This is the major component used in plastic products, including bottles, containers, storage items, and food serving ware. Exposure of BPA happens through oral, respiratory, transdermal routes and eye contact. As an EDC, BPA disrupts hormonal binding, leading to various health problems, such as cancers, reproductive abnormalities, metabolic syndrome, immune dysfunction, neurological effects, cardiovascular problems, respiratory issues, and obesity. BPA mimics the hormone estrogen but exhibits a weak affinity for estrogen receptors. This weak binding affinity triggers multiple cell death pathways, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, across different cell types. Numerous clinical, in-vitro, and in-vivo experiments have demonstrated that BPA exposure results in unfavorable health effects. This review highlights the mechanisms of cell death pathways initiated through BPA exposure and the associated negative health consequences. The extensive use of BPA and its frequent detection in environmental and biological models underscore the urgent need for further investigation into its effects and the development of safe alternatives. Addressing the health risks posed by BPA involves a comprehensive approach that includes reducing exposure and finding novel substitutes to lessen its detrimental impact on humans.
Collapse
Affiliation(s)
- Yukta S
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Milan K L
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Tokyo, Japan
| | - S Sahabudeen
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Tokyo, Japan
| | - K M Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
36
|
Hao B, Lin S, Liu H, Xu J, Chen L, Zheng T, Zhang W, Dang Y, Reiter RJ, Li C, Zhai H, Xia Q, Fan L. Baicalein tethers CD274/PD-L1 for autophagic degradation to boost antitumor immunity. Autophagy 2025; 21:917-933. [PMID: 39710370 PMCID: PMC12013432 DOI: 10.1080/15548627.2024.2439657] [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: 12/13/2023] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
Immune checkpoint inhibitors, especially those targeting CD274/PD-L1yield powerful clinical therapeutic efficacy. Thoughmuch progress has been made in the development of antibody-basedCD274 drugs, chemical compounds applied for CD274degradation remain largely unavailable. Herein,baicalein, a monomer of traditional Chinese medicine, isscreened and validated to target CD274 and induces itsmacroautophagic/autophagic degradation. Moreover, we demonstrate thatCD274 directly interacts with MAP1LC3B (microtubule associatedprotein 1 light chain 3 beta). Intriguingly, baicalein potentiatesCD274-LC3 interaction to facilitate autophagic-lysosomal degradationof CD274. Importantly, targeted CD274. degradation via baicaleininhibits tumor development by boosting T-cell-mediated antitumorimmunity. Thus, we elucidate a critical role of autophagy-lysosomalpathway in mediating CD274 degradation, and conceptually demonstratethat the design of a molecular "glue" that tethers the CD274-LC3interaction is an appealing strategy to develop CD274 inhibitors incancer therapy.Abbreviations: ATTECs: autophagy-tethering compounds; AUTACs: AUtophagy-TArgeting Chimeras; AUTOTACs: AUTOphagy-TArgeting Chimeras; AMPK: adenosine 5'-monophosphate (AMP)-activated protein kinase; BiFC: bimolecular fluorescence complementation; BafA1: bafilomycin A1; CD274/PD-L1/B7-H1: CD274 molecule; CQ: chloroquine; CGAS: cyclic GMP-AMP synthase; DAPI: 4'6-diamino-2-phenylindole; FITC: fluorescein isothiocyanate isomer; GFP: green fluorescent protein; GZMB: granzyme B; IHC: immunohistochemistry; ICB: immune checkpoint blockade; KO: knockout; KD: equilibrium dissociation constant; LYTAC: LYsosome-TArgeting Chimera; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MST: microscale thermophoresis; NFAT: nuclear factor of activated T cells; NFKB/NF-kB: nuclear factor kappa B; NSCLC: non-small-cell lung cancer; PDCD1: programmed cell death 1; PROTACs: PROteolysis TArgeting Chimeras; PRF1: perforin 1; PE: phosphatidylethanolamine; PHA: phytohemagglutinin; PMA: phorbol 12-myristate 13-acetate; STAT: signal transducer and activator of transcription; SPR: surface plasmon resonance; TILs: tumor-infiltrating lymphocyte; TME: tumor microenvironment.
Collapse
Affiliation(s)
- Bingjie Hao
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumeng Lin
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haipeng Liu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junfang Xu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li Chen
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tiansheng Zheng
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen Zhang
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yifang Dang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Chaoqun Li
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong Zhai
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Xia
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lihong Fan
- Institute of Energy Metabolism and Health, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
37
|
Bai H, Xi G, Cheng Y. Prostaglandin G/H synthase 1 promotes thrombosis in atrial fibrillation through modulation of platelet activation, macrophage infiltration, inflammation, and autophagy inhibition. Acta Cardiol 2025; 80:254-265. [PMID: 39963884 DOI: 10.1080/00015385.2025.2467009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/15/2025] [Accepted: 02/09/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Prostaglandin G/H synthase 1 (PTGS1) is known to regulate platelet function and inflammation. However, its role in atrial fibrillation (AF)-related thrombosis is not well understood. This study investigates the role of PTGS1 in AF-associated thrombus formation and its underlying mechanisms. METHODS Left atrial appendage (LAA) tissues were collected from 48 patients undergoing valve replacement surgery, divided into three groups: sinus rhythm (SR), AF with thrombus [AF (+) T (+)], and AF without thrombus [AF (+) T (-)]. PTGS1 expression, platelet activation markers (MPA, sCD40L, and d-dimer), macrophage phenotypes (M1 and M2), inflammatory cytokines (IL-1β, TNF-α, IL-6), and autophagy-related proteins (LC3II and p62) were assessed. Furthermore, the effect of PTGS1 manipulation on autophagy in endocardial endothelial cells (EECs) was examined using cell transfection experiments. RESULTS PTGS1 expression was significantly higher in LAA tissues of AF (+) T (+) patients compared to AF (+) T (-) and SR groups. It was positively correlated with reduced LAA emptying velocity (LAAEV), higher CHA2DS2-VASc scores, and elevated platelet activation markers (MPA, sCD40L, and d-dimer). Data also showed increased M1 macrophage infiltration and higher pro-inflammatory cytokines (IL-1β, TNF-α, IL-6) in AF (+) T (+) patients, with PTGS1 expression strongly linked to these markers. Furthermore, PTGS1 overexpression inhibited autophagy in EECs by decreasing LC3II/LC3I ratio and increasing p62 levels, while PTGS1 knockdown promoted autophagy, protecting against endothelial dysfunction. CONCLUSIONS PTGS1 is overexpressed in AF patients with thrombosis and may play an important role in promoting thrombus formation through enhanced platelet activation, inflammation, and inhibition of autophagy.
Collapse
Affiliation(s)
- Hao Bai
- Department of Ultrasound Diagnosis, Anting Hospital, Shanghai, China
| | - Guiyang Xi
- Department of Ultrasound Diagnosis, Anting Hospital, Shanghai, China
| | - Yangyang Cheng
- Department of Ultrasound Diagnosis, Anting Hospital, Shanghai, China
| |
Collapse
|
38
|
Elkhalil A, Whited A, Ghose P. SQST-1/p62-regulated SKN-1/Nrf mediates a phagocytic stress response via transcriptional activation of lyst-1/LYST. PLoS Genet 2025; 21:e1011696. [PMID: 40315422 PMCID: PMC12068719 DOI: 10.1371/journal.pgen.1011696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 05/12/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025] Open
Abstract
Cells may be intrinsically fated to die to sculpt tissues during development or to maintain homeostasis. Cells can also die in response to various stressors, injury or pathological conditions. Additionally, cells of the metazoan body are often highly specialized with distinct domains that differ both structurally and with respect to their neighbors. Specialized cells can also die, as in normal brain development or pathological states and their different regions may be eliminated via different programs. Clearance of different types of cell debris must be performed quickly and efficiently to prevent autoimmunity and secondary necrosis of neighboring cells. Moreover, all cells, including those programmed to die, may be subject to various stressors. Some largely unexplored questions include whether predestined cell elimination during development could be altered by stress, if adaptive stress responses exist and if polarized cells may need compartment-specific stress-adaptive programs. We leveraged Compartmentalized Cell Elimination (CCE) in the nematode C. elegans to explore these questions. CCE is a developmental cell death program whereby three segments of two embryonic polarized cell types are eliminated differently. We have previously employed this in vivo genetic system to uncover a cell compartment-specific, cell non-autonomous clearance function of the fusogen EFF-1 in phagosome closure during corpse internalization. Here, we introduce an adaptive response that serves to aid developmental phagocytosis as a part of CCE during stress. We employ a combination of forward and reverse genetics, CRISPR/Cas9 gene editing, stress response assays and advanced fluorescence microscopy. Specifically, we report that, under heat stress, the selective autophagy receptor SQST-1/p62 promotes the nuclear translocation of the oxidative stress-related transcription factor SKN-1/Nrf via negative regulation of WDR-23. This in turn allows SKN-1/Nrf to transcribe lyst-1/LYST (lysosomal trafficking associated gene) which subsequently promotes the phagocytic resolution of the developmentally-killed internalized cell even under stress conditions.
Collapse
Affiliation(s)
- Aladin Elkhalil
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Alec Whited
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Piya Ghose
- The University of Texas at Arlington, Arlington, Texas, United States of America
| |
Collapse
|
39
|
Khodadadi H, Łuczyńska K, Winiarczyk D, Leszczyński P, Taniguchi H. NFE2L1 as a central regulator of proteostasis in neurodegenerative diseases: interplay with autophagy, ferroptosis, and the proteasome. Front Mol Neurosci 2025; 18:1551571. [PMID: 40375958 PMCID: PMC12078313 DOI: 10.3389/fnmol.2025.1551571] [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: 12/25/2024] [Accepted: 03/14/2025] [Indexed: 05/18/2025] Open
Abstract
Maintaining proteostasis is critical for neuronal health, with its disruption underpinning the progression of neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's diseases. Nuclear Factor Erythroid 2-Related Factor 1 (NFE2L1) has emerged as a key regulator of proteostasis, integrating proteasome function, autophagy, and ferroptosis to counteract oxidative stress and protein misfolding. This review synthesizes current knowledge on the role of NFE2L1 in maintaining neuronal homeostasis, focusing on its mechanisms for mitigating proteotoxic stress and supporting cellular health, offering protection against neurodegeneration. Furthermore, we discuss the pathological implications of NFE2L1 dysfunction and explore its potential as a therapeutic target. By highlighting gaps in the current understanding and presenting future research directions, this review aims to elucidate NFE2L1's role in advancing treatment strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Hossein Khodadadi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Kamila Łuczyńska
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, Warsaw, Poland
| | - Dawid Winiarczyk
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Paweł Leszczyński
- Department of Stem Cell Bioengineering Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- African Genome Center, University Mohammed VI Polytechnic (UM6P), Ben Guerir, Morocco
| |
Collapse
|
40
|
Hossain MM, Mishra AK, Yadav AK, Akanksha, Ismail M, Sata TN, Sah AK, Al Mohit A, Venugopal SK. MicroRNA-122 regulates inflammatory and autophagic proteins by downregulating pyruvate kinase M2 in non-alcoholic fatty liver disease. Mol Cell Biochem 2025; 480:3067-3078. [PMID: 39630362 DOI: 10.1007/s11010-024-05174-y] [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: 05/04/2024] [Accepted: 11/20/2024] [Indexed: 05/03/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the serious global health concerns, leading to non-alcoholic steatohepatitis (NASH), and to hepatocellular carcinoma (HCC). Despite its prevalence, the molecular mechanisms regulating NAFLD progression remain elusive. The present study aims to determine role of microRNA-122-mediated regulation of pyruvate kinase M2 (PKM2) on regulating inflammatory and autophagic proteins during the pathogenesis of NAFLD. Huh7 cells were incubated with free fatty acids (FFAs) or transfected with single guide RNA to PKM2 containing CRISPR-Cas9 system or miR-122 for up to 72 h. C57BL/6 mice were fed with sham-operated control, choline sufficient L-amino acid defined (CSAA) or choline-deficient L-amino acid defined (CDAA) diet for 6, 18, 32 and 54 weeks. The RNA or protein was isolated from the Huh7 cells and the liver tissue of the mice. RT-PCR was performed for miR-122 expression and Western blots were performed for PKM2, iNOS, COX2, Beclin-1, Atg7 and LC3-II. FFAs induced the expression of PKM2, iNOS and COX2, while decreased the expression of miR-122, Beclin-1, Atg7 and LC3-II. Overexpression of miR-122 resulted in decreased PKM2, iNOS and COX2 and increased Beclin-1, Atg7 and LC3-II. Silencing of PKM2 led to decreased iNOS and COX2 and increased Beclin-1, Atg7 and LC3-II. In CDAA fed-mice, there was a significant increase in PKM2, iNOS and COX2 and decreased miR-122, Beclin-1, Atg7 and LC3-II. The data showed that FFAs downregulated miR-122 expression, which resulted in the upregulation of PKM2, which in turn upregulated inflammatory proteins and downregulated autophagic proteins during the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Md Musa Hossain
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
| | - Amit K Mishra
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New Delhi, 110068, USA
| | - Ajay K Yadav
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Akanksha
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
| | - Md Ismail
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
| | - Teja Naveen Sata
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
| | - Amrendra K Sah
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
| | - Abdullah Al Mohit
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India
| | - Senthil K Venugopal
- Faculty of Life Sciences and Biotechnology, South Asian University, Rajpur Road, Maidan Garhi, New Delhi, India.
| |
Collapse
|
41
|
Yi Z, Liu J, Shen L, Hu Y. mTOR and autophagy in acute lung injury pathogenesis and therapeutic potential. J Thorac Dis 2025; 17:2679-2692. [PMID: 40400934 PMCID: PMC12090145 DOI: 10.21037/jtd-24-1817] [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: 10/24/2024] [Accepted: 02/28/2025] [Indexed: 05/23/2025]
Abstract
Acute lung injury (ALI) poses a significant clinical challenge due to its high morbidity and mortality rates. Current treatment options are limited in their efficacy, necessitating the exploration of novel therapeutic targets. The mammalian target of rapamycin (mTOR), a crucial regulator of various cellular processes, has been implicated in the pathogenesis of ALI. Autophagy, a tightly regulated cellular degradation process controlled by mTOR, plays a pivotal role in the pathogenesis of ALI and cellular homeostasis. Mounting evidence also suggests that the mTOR pathway and autophagy play crucial roles in the pathogenesis and regulation of ALI. Herein, we reviewed the current understanding of how mTOR signaling and autophagy intersect in the context of ALI, with a focus on their roles across different cell types. This analysis highlights their dual roles in either promoting pulmonary injury or providing protection, depending on the specific cell types and different ALI models. Insights into the intricate balance between mTOR-mediated pathways and autophagic responses provide a foundation for developing targeted therapeutic strategies aimed at alleviating ALI through the modulation of these pathways. This review underscores the therapeutic potential of targeting mTOR and autophagy, presenting innovative and promising approaches for improving the clinical management and outcomes of ALI.
Collapse
Affiliation(s)
- Zihan Yi
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiahao Liu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lanying Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Hu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Luo R, Wang Z, Xu F, Xie K. Dexmedetomidine improve lung inflammation by regulating autophagy and apoptosis of CD4+ T cell via AMPK/mTOR signaling. Mol Immunol 2025; 183:1-11. [PMID: 40311186 DOI: 10.1016/j.molimm.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/11/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVES To investigate the protective effect and potential mechanism of dexmedetomidine (Dex) in acute lung injury (ALI). MATERIALS AND METHODS C57BL/6 mice and EL-4 cells were used for in vivo and in vitro studies, respectively. Cecal ligation and puncture (CLP) method was used to prepare an acute lung injury model. After dexmedetomidine intervention, tissue and cell samples were collected to evaluate and measure the severity of lung damage, the proportion of Treg cells, the expression of autophagy-related protein levels and AMPK/mTOR pathways. RESULTS Dex reduced lung damage, and IL-17a, MPO positive cells in the lung, decreased the levels of pro-inflammatory cytokines, and restrain autophagy and apoptosis via the activation of the AMPK/mTOR pathway and increase of the proportion of Tregs. CONCLUSIONS Dex can inhibit the levels of autophagy and apoptosis, increase the proportion of Treg cells, and reduce CLP induced acute lung injury through regulating AKMP/MTOR pathway.
Collapse
Affiliation(s)
- Renjie Luo
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, Chongqing, China
| | - Zhao Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Ke Xie
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Sepsis Diagnosis and Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
43
|
Liu J, Ge P, Luo Y, Sun Z, Luo X, Li H, Pei B, Xun L, Zhang X, Jiang Y, Wen H, Liu J, Yang Q, Ma S, Chen H. Decoding TMAO in the Gut-Organ Axis: From Biomarkers and Cell Death Mechanisms to Therapeutic Horizons. Drug Des Devel Ther 2025; 19:3363-3393. [PMID: 40322030 PMCID: PMC12049683 DOI: 10.2147/dddt.s512207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
The gut microbiota and its metabolites are bi-directionally associated with various human illnesses, which has received extensive attention. Trimethylamine N-oxide (TMAO) is a gut microbiota metabolite produced in the liver, which may serve the role of an "axis" connecting the gut and host organs. TMAO levels are significantly higher in the blood of individuals with cardiovascular, renal, neurological, and metabolic diseases. Endothelial cells are crucial for regulating microcirculation and maintaining tissue and organ barriers and are widely recognized as target cells for TMAO. TMAO not only induces endothelial dysfunction but also acts on various cell types, such as endothelial cells, epithelial cells, vascular smooth muscle cells, nerve cells, and pancreatic cells, triggering multiple cell death mechanisms, including necrosis and programmed cell death, thereby influencing host health. This paper thoroughly covers the origins, production, and metabolic pathways of TMAO, emphasizing its importance in the early detection and prognosis of human diseases in the "Gut-Organ" axis, as well as its mechanisms of influence on human diseases, particularly the cross-talk with cell death. Furthermore, we cover recent advances in treating human diseases by regulating gut microbiota structure and enzyme activity to influence TMAO metabolism and reduce TMAO levels, including the use of probiotics, prebiotics, antibiotics, anti-inflammatory drugs, antiplatelet drugs, hypoglycemic drugs, lipid-lowering drugs, and natural products.
Collapse
Affiliation(s)
- Jie Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Peng Ge
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Yalan Luo
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Zhenxuan Sun
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Xinyu Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Huijuan Li
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Boliang Pei
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Lu Xun
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Xuetao Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Yunfei Jiang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Haiyun Wen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Qi Yang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, People’s Republic of China
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| |
Collapse
|
44
|
Horcas-Nieto JM, Rios-Ocampo WA, Langelaar-Makkinje M, de Boer R, Gerding A, Chornyi S, Martini IA, Wolters JC, Wanders RJA, Waterham HR, Van der Klei IJ, Bandsma RHJ, Jonker JW, Bakker BM. Docosahexaenoic acid prevents peroxisomal and mitochondrial protein loss in a murine hepatic organoid model of severe malnutrition. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167849. [PMID: 40306148 DOI: 10.1016/j.bbadis.2025.167849] [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: 06/05/2024] [Revised: 02/20/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Acute and chronic exposure of cells to low amino acid conditions have been shown to lead to a reduction in hepatic peroxisomal and mitochondrial content. There is limited understanding of the underlying mechanisms behind this loss, but data suggests degradation through autophagy. Both organelles play a key role in fatty acid metabolism, which may explain why dysfunction in either one of them might lead to hepatic steatosis. METHODS Using a previously established murine hepatic organoid model of severe malnutrition, we characterized the effects of prolonged amino-acid restriction on peroxisomal and mitochondrial protein levels and on autophagic flux. To do so, we developed concatemers of 13C-labelled peptide standards for quantification of over 50 different peroxisomal proteins. To assess the autophagic flux, we transduced hepatic organoids with a GFP-LC3-RFP-LC3ΔG probe. Finally, the effect of PPAR-α activation on peroxisomal loss was determined with various agonists. RESULTS Prolonged (96 h) amino-acid restriction led to a more severe loss of peroxisomes than a 48 h restriction, and with a substantial induction of autophagic flux. This was accompanied by accumulation of intracellular triglycerides, loss of mitochondrial and peroxisomal proteins, and loss of peroxisomal functionality. While PPAR-α agonists WY-14643 and linoleic acid (LA) had no effect, docosahexaenoic acid (DHA) supplementation partly prevented peroxisomal and mitochondrial loss under amino-acid restricted conditions and partly inhibited autophagy. DISCUSSION The potential of DHA to prevent loss of peroxisomes and mitochondrial functions in low protein diets and severe malnutrition warrants further causal and translational testing in preclinical models and clinical trials, including its use as nutritional supplement.
Collapse
Affiliation(s)
- José M Horcas-Nieto
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - W Alfredo Rios-Ocampo
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Rinse de Boer
- Molecular Cell Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Albert Gerding
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Serhii Chornyi
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ingrid A Martini
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Justina C Wolters
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Interfaculty Mass Spectrometry Center, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ida J Van der Klei
- Molecular Cell Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, the Netherlands
| | - Robert H J Bandsma
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada
| | - Johan W Jonker
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Barbara M Bakker
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands.
| |
Collapse
|
45
|
Tu WC, He YK, Wang DW, Ming SX, Zhao Y. Progranulin enhances M2 macrophage polarization and renal fibrosis by modulating autophagy in chronic kidney disease. Cell Mol Life Sci 2025; 82:186. [PMID: 40293508 PMCID: PMC12037463 DOI: 10.1007/s00018-025-05716-7] [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: 02/13/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a prevalent global health issue characterized by progressive renal dysfunction and fibrosis, often leading to end-stage renal failure. Renal fibrosis, a hallmark of CKD, is driven by complex immune responses, including macrophage polarization and inflammatory signaling pathways. Progranulin (PGRN), a glycoprotein involved in inflammation and tissue repair, has emerged as a key regulator in various fibrotic diseases. However, the precise role of PGRN in macrophage polarization and renal fibrosis in CKD remains unclear and warrants further investigation. METHODS Renal tissue samples from CKD patients and unilateral ureteral obstruction (UUO)-induced mice were analyzed using immunohistochemistry, immunofluorescence, Western blotting, and qRT-PCR to assess fibrosis, macrophage infiltration, and key markers of autophagy and inflammation. Recombinant PGRN (rPGRN) was administered in vivo to assess its effects on renal fibrosis, macrophage polarization, and autophagic flux. To evaluate the role of PGRN, PGRN knockout (PGRN-/-) mice were also utilized. The effects of PGRN on autophagic flux and mitochondrial dynamics were studied using mCherry-GFP-LC3 dual-labeling, and macrophage polarization was analyzed by flow cytometry and cytokine profiling. RESULTS PGRN expression is significantly elevated in CKD patients and UUO mice and is associated with increased macrophage infiltration and renal fibrosis. rPGRN administration in vivo aggravated fibrosis and promoted M2 macrophage polarization. In contrast, PGRN-/- mice showed reduced renal fibrosis, significantly reduced collagen deposition, and reduced expression of pro-fibrotic cytokines. In addition, the mitochondrial function of PGRN-/- renal fibrosis mice was improved, the mtDNA content of mouse kidney tissue was increased, the results of electron microscopy showed that the mitochondrial structure was relatively normal, the mitochondrial biogenesis related genes PGC1α, TOMM20 and Fis1 were up-regulated, and the levels of MFN2 and Drp1 were significantly reduced. In addition, autophagy related gene LC3 was decreased and P62 protein level was increased in PGRN-/- model mice. Mechanically, PGRN interacts with autophagy related proteins ATG5 and ATG12 to regulate autophagy flux through the PI3K-Akt signaling pathway and promote the polarization of M2 macrophages. CONCLUSION PGRN plays a critical role in driving renal fibrosis by regulating macrophage polarization, autophagy, and mitochondrial dynamics. Our findings suggest that PGRN exacerbates CKD progression by promoting M2 macrophage polarization and disrupting autophagic processes, highlighting PGRN as a potential therapeutic target for the treatment of CKD and renal fibrosis.
Collapse
Affiliation(s)
- Wei-Chao Tu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, No. 999 Hope Road, Jiading District, Shanghai, 201800, China
| | - Yi-Kun He
- Department of Rheumatism, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Pudong, 201203, China
| | - Da-Wei Wang
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, No. 999 Hope Road, Jiading District, Shanghai, 201800, China
| | - Shao-Xiong Ming
- Department of Urology, Shanghai Changhai Hospital, No.168 Changhai Rd, Shanghai, 200433, China.
| | - Yang Zhao
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, No. 999 Hope Road, Jiading District, Shanghai, 201800, China.
| |
Collapse
|
46
|
Salem MA, Khalil HMA, Manaa EG, Bass AKA, Osama N, Samaka RM, Ibrahim MT, Hamdan DI. Antioxidant Potential of Selected Apiaceae Plant Extracts: A Study Focused on the Chemical Composition and Neuroprotective Effect of Coriandrum sativum L. Extract Against Lead (Pb)-Induced Neurotoxicity in Rats. Biol Trace Elem Res 2025:10.1007/s12011-025-04627-9. [PMID: 40261543 DOI: 10.1007/s12011-025-04627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Lead is a common environmental pollutant. It can affect several body systems including the central nervous system (CNS). Lead can disrupt the nervous system by different mechanisms including oxidative stress, inflammation, disruption of neurotransmission, and aberrant autophagy. Apiaceous species have been used traditionally as food flavoring and medicine, representing a rich source of bioactive compounds. In the current study, the antioxidant power of four Apiaceous extracts (Foeniculum vulgare L., Pimpinella anisum L., Coriandrum sativum L., and Cuminum cyminum L.) was evaluated. Additionally, the metabolite profiles of the selected species were comprehensively investigated by untargeted liquid chromatography electrospray ionization tandem mass spectrometry (LC/ESI-MS/MS) coupled to chemometry. Coriander (Coriandrum sativum L.) extract showed the highest radical scavenging activity and reducing power. Coriander was further subjected to in vivo evaluation of its protective effect against Lead (Pb)-induced neurotoxicity. Administration of coriander extracts improved the short- and long-term memory performance and decreased hippocampal Pb content in Pb-intoxicated rats. Moreover, it attenuated hippocampal oxidative stress, neurochemical changes, and exhibited anti-inflammatory effect in the hippocampal tissue. Further, coriander extracts attenuated Pb inhibitory effect on the mammalian target of Rapamycin (mTORC1) pathway resulting in upregulation of Phospho-p70 S6 Kinase (P-P70S6K) and Phospho-S6 Ribosomal Protein (PS6) and downregulation of Beclin-1. Additionally, some selected coriander ingredients were subjected to molecular docking to examine their regulatory effect on mTORC-1 and IκB kinase complex (Ikk-β). The present findings highlight the future pharmaceutical utilization of coriander extract as valuable source of phenolic compounds that can be used as antioxidant, anti-inflammatory, and neuroprotective agents against Pb-induced neurotoxicity.
Collapse
Affiliation(s)
- Mohamed A Salem
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University, Gamal Abd El Nasr St., Shibin Elkom, Menoufia, 32511, Egypt
- The BioActives Lab, Biological and Environment Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Heba M A Khalil
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
- Faculty of Veterinary medicine, King Salman International University, South Sinai, Ras Sudr, Egypt
| | - Eman G Manaa
- Department of Pharmacognosy, Faculty of Pharmacy, Alsalam University, Al Gharbiyah, Egypt
| | - Amr K A Bass
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia National University, Km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| | - Nada Osama
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, Gamal Abd El Nasr St., Shibin Elkom, Menoufia, 32511, Egypt.
| | - Rehab M Samaka
- Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Magda T Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, 11785, Egypt
| | - Dalia I Hamdan
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University, Gamal Abd El Nasr St., Shibin Elkom, Menoufia, 32511, Egypt
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia National University, Km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| |
Collapse
|
47
|
Ma R, Cui Y, Yu SJ, Pan YY, He JF, Wang YY, Wang JL, Wang XY, Bai XF, Zhang H, Yang SS, Zhang Q. The glucose metabolism reprogramming of yak Sertoli cells under hypoxia is regulated by autophagy. BMC Genomics 2025; 26:385. [PMID: 40251498 PMCID: PMC12007286 DOI: 10.1186/s12864-025-11497-x] [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: 02/06/2024] [Accepted: 03/18/2025] [Indexed: 04/20/2025] Open
Abstract
Hypoxia often has negative effects on testis development and spermatogenesis of mammals. Plateau yaks have lived in the hypoxia environment for generations, but have ensured testicular function, which is closely related to their unique hypoxia response mechanism. Glucose metabolic reprogramming is an important way for cells to respond to stressful environments, especially the metabolite lactate, which is the energy basis for the development and differentiation of germ cells. In this study, hypoxia (5% O2) effectively promoted yak Sertoli cell proliferation and decreased autophagy and apoptosis. It was found that the cells showed good hypoxic adaptation. Metabolomics results showed that glucose metabolism was enhanced in yak Sertoli cells in response to hypoxia, and 13 glucose metabolites were increased, including the production and transport level of lactic acid (LA), which may have changed the pentose phosphate metabolic pathway of cells, these changes are conducive to support the glucose metabolism balance of cells under hypoxia. Crucially, when autophagy is activated under hypoxia, GLUT3, GLUT8, and MCT4 proteins are degraded, while GLUT1 and MCT1 are not affected, suggesting that autophagy may achieve glucose metabolic reprogramming by selectively regulating the expression of functional factors of glucose metabolism, which is conducive to energy intake and spermatogenesis in testis of yaks.
Collapse
Affiliation(s)
- Rui Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China.
| | - Si-Jiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China.
| | - Yang-Yang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Jun-Feng He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Ya-Ying Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Jing-Lei Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Xiao-Yan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Xue-Feng Bai
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Hui Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Shan-Shan Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| | - Qian Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
- Gansu Province Livestock Embryo Engineering Research Center, Lanzhou, 730070, China
| |
Collapse
|
48
|
Huang H, Fang L, Zhu C, Lv J, Xu P, Chen Z, Zhang Z, Wang J, Wang W, Xu Z. YBX1 promotes 5-Fluorouracil resistance in gastric cancer via m5C-dependent ATG9A mRNA stabilization through autophagy. Oncogene 2025:10.1038/s41388-025-03411-2. [PMID: 40251390 DOI: 10.1038/s41388-025-03411-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
5-Fluorouracil (5-FU) is a first-line chemotherapeutic agent for advanced gastric cancer (GC). However, its clinical efficacy is often undermined by the development of chemoresistance. Aberrant activation of oncogenic pathways, including autophagy, has been implicated in 5-FU resistance. Epigenetic modifications, such as 5-methylcytosine (m5C), are also recognized to modulate autophagy and contribute to chemoresistance, though the underlying molecular mechanisms remain poorly understood. In this study, we discovered that YBX1, an m5C reader protein, was significantly upregulated in 5-FU-resistant GC cell lines and patient tissues. Both in vitro and in vivo experiments demonstrated that YBX1 promoted autophagy in GC cells, thereby enhancing 5-FU resistance. Mechanistically, the transcription factor MAZ was found to bind to the YBX1 promoter, driving its transcriptional upregulation. YBX1, in turn, stabilized ATG9A mRNA via NSUN2-mediated m5C modification, thereby enhancing autophagic activity and conferring chemoresistance. Clinically, elevated YBX1 expression correlated with poor prognosis in patients with advanced GC undergoing 5-FU-based chemotherapy. These findings establish YBX1 as a key regulator of autophagy and 5-FU resistance in GC and highlight its potential as a novel therapeutic target for overcoming 5-FU resistance.
Collapse
Affiliation(s)
- Hongxin Huang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Lang Fang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chuming Zhu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jialun Lv
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Penghui Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zetian Chen
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhijun Zhang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jihuan Wang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Weizhi Wang
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Zekuan Xu
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- Institute for Gastric Cancer Research, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
49
|
Tian J, Mao Y, Liu D, Li T, Wang Y, Zhu C. Mitophagy in Brain Injuries: Mechanisms, Roles, and Therapeutic Potential. Mol Neurobiol 2025:10.1007/s12035-025-04936-z. [PMID: 40237948 DOI: 10.1007/s12035-025-04936-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Mitophagy is an intracellular degradation pathway crucial for clearing damaged or dysfunctional mitochondria, thereby maintaining cellular homeostasis and responding to various brain injuries. By promptly removing damaged mitochondria, mitophagy protects cells from further harm and support cellular repair and recovery after injury. In different types of brain injury, mitophagy plays complex and critical roles, from regulating the balance between cell death and survival to influencing neurological recovery. This review aims to deeply explore the role and mechanism of mitophagy in the context of brain injuries and uncover how mitophagy regulates the brain response to injury and its potential therapeutic significance. It emphasizes mitophagy's potential in treating brain injuries, including reducing cell damage, promoting cell recovery, and improving neurological function, thus opening new perspectives and directions for future research and clinical applications.
Collapse
Affiliation(s)
- Jiayu Tian
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yanna Mao
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Dandan Liu
- Department of Electrocardiogram, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Tao Li
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China.
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China.
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscienceand , Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530, Göteborg, Sweden.
| |
Collapse
|
50
|
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
|