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
|
Xu NN, Wen YC, Pan J, Shu F, Qu JX, Qi XF, Tang J. Activated mTORC1 signaling pathway aggravates cisplatin induced oxidative damage by inhibiting autophagy in mouse cochlear hair cells. Neuropharmacology 2025; 272:110433. [PMID: 40147638 DOI: 10.1016/j.neuropharm.2025.110433] [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: 11/22/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Platinum-based antitumor drugs, such as cisplatin and carboplatin, are well-known for their severe ototoxicity. The ototoxic effects of these drugs are primarily attributed to oxidative stress induced damage within cochlear hair cells (HCs), leading to cell death and subsequent irreversible hearing loss. Over the past decade, studies have demonstrated that upregulating autophagy levels in HCs can greatly alleviate the death of cochlear HCs as part of the oxidative damage induced by ototoxic drugs. However, the molecular mechanisms by which platinum-based drugs affect autophagy and ultimately lead to HCs death remain unclear. In the present study, we investigated the effects of cisplatin on the mTOR signaling pathway, a critical regulator of autophagy, in cochlear explants of mice. Our results indicated that while cisplatin enhances autophagy activity initially, it also activates mTOR Complex1 (mTORC1) within HCs. The persistent activation of mTORC1 inhibits autophagy in HCs, resulting in the accumulation of reactive oxygen species and leading to cell death. Further pharmacological experiments confirmed the protective role of rapamycin, a specific mTORC1 inhibitor, highlighting the importance of autophagy in combating cisplatin-induced ototoxicity. Our findings suggest that modulating the mTOR signaling pathway to regulate autophagy could be an effective strategy for preventing cisplatin-induced ototoxic damage.
Collapse
Affiliation(s)
- Na-Na Xu
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yin-Chuan Wen
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518000, China
| | - Jing Pan
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Fan Shu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jia-Xi Qu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xiao-Fei Qi
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518000, China.
| | - Jie Tang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China; Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Chen Y, Deng H, Zhang N. Autophagy-targeting modulation to promote peripheral nerve regeneration. Neural Regen Res 2025; 20:1864-1882. [PMID: 39254547 PMCID: PMC11691477 DOI: 10.4103/nrr.nrr-d-23-01948] [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: 11/27/2023] [Revised: 02/22/2024] [Accepted: 03/29/2024] [Indexed: 09/11/2024] Open
Abstract
Nerve regeneration following traumatic peripheral nerve injuries and neuropathies is a complex process modulated by diverse factors and intricate molecular mechanisms. Past studies have focused on factors that stimulate axonal outgrowth and myelin regeneration. However, recent studies have highlighted the pivotal role of autophagy in peripheral nerve regeneration, particularly in the context of traumatic injuries. Consequently, autophagy-targeting modulation has emerged as a promising therapeutic approach to enhancing peripheral nerve regeneration. Our current understanding suggests that activating autophagy facilitates the rapid clearance of damaged axons and myelin sheaths, thereby enhancing neuronal survival and mitigating injury-induced oxidative stress and inflammation. These actions collectively contribute to creating a favorable microenvironment for structural and functional nerve regeneration. A range of autophagy-inducing drugs and interventions have demonstrated beneficial effects in alleviating peripheral neuropathy and promoting nerve regeneration in preclinical models of traumatic peripheral nerve injuries. This review delves into the regulation of autophagy in cell types involved in peripheral nerve regeneration, summarizing the potential drugs and interventions that can be harnessed to promote this process. We hope that our review will offer novel insights and perspectives on the exploitation of autophagy pathways in the treatment of peripheral nerve injuries and neuropathies.
Collapse
Affiliation(s)
- Yan Chen
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Hongxia Deng
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Nannan Zhang
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
4
|
Ge L, Ma J, Xu J, Wang B, Adil A, Xu H. The mechanism of lncRNA PVT1 targeting the miR-30a/Beclin-1 axis to mediate ventricular remodeling in spontaneously hypertensive rats. Cell Signal 2025; 130:111650. [PMID: 39923929 DOI: 10.1016/j.cellsig.2025.111650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Hypertension poses a great health threat globally. We probed the mechanisms of long non-coding RNA plasmacytoma variant translocation 1 (lncRNA PVT1) mediating ventricular remodeling (VR) in spontaneously hypertensive rats (SHR). METHODS PVT1 was down-regulated or miR-30a was inhibited in SHR in vivo. Hypertensive injury model was established in vitro. VR, fibrosis and autophagy-related indicators were detected by echocardiography, HE/WGA/Masson staining, ELISA, and immunohistochemistry. Cell viability, fibrosis markers, autophagy-related markers, and lncRNA PVT1 and miR-30a levels were assessed. Interactions between PVT1, Beclin-1 and miR-30a were verified. RESULTS PVT1 was up-regulated in myocardial tissues of SHR. PVT1 knockdown alleviated VR and myocardial fibrosis (MF) in SHR, as evidenced by decreased systolic blood pressure, left ventricular end-systolic diameter, left ventricular end-systolic diameter, and heart weight index, boosted left ventricular fractional shortening and left ventricular ejection fraction, abated inflammatory infiltration of myocardial tissues, decreased myocardial hypertrophy and interstitial fibrosis, reduced serum angiotensin II (Ang II) and atrial natriuretic peptide, and downregulated collagen I, collagen II, α-smooth muscle actin, and fibronectin protein. PVT1 knockdown down-regulated Beclin 1 and LC3B-II/LC3B-I and up-regulated p62 protein. In vitro, PVT1 knockdown improved fibrosis by inhibiting Ang II-induced cardiomyocyte autophagy. PVT1 acted as a competitive endogenous RNA to competitively bind to miR-30a to target Beclin-1 expression. PVT1 targeted the miR-30a/Beclin-1 axis to mediate autophagy to affect VR and MF in SHR. CONCLUSIONS LncRNA PVT1 promotes cellular autophagy by targeting the miR-30a/Beclin-1 axis, thereby promoting VR and MF in SHR. Knockdown of lncRNA PVT1 attenuates VR and MF in SHR.
Collapse
Affiliation(s)
- Li Ge
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Jianjun Ma
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Jingxuan Xu
- School of Optometry and Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Bo Wang
- Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Abdusalam Adil
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Hongfeng Xu
- Department of Hypertension, The Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
5
|
Zhao P, Tian R, Song D, Zhu Q, Ding X, Zhang J, Cao B, Zhang M, Xu Y, Fang J, Tan J, Yi C, Xia H, Liu W, Zou W, Sun Q. Rab GTPases are evolutionarily conserved signals mediating selective autophagy. J Cell Biol 2025; 224:e202410150. [PMID: 40197538 PMCID: PMC11977514 DOI: 10.1083/jcb.202410150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/31/2024] [Accepted: 01/21/2025] [Indexed: 04/10/2025] Open
Abstract
Selective autophagy plays a crucial role in maintaining cellular homeostasis by specifically targeting unwanted cargo labeled with "autophagy cues" signals for autophagic degradation. In this study, we identify Rab GTPases as a class of such autophagy cues signals involved in selective autophagy. Through biochemical and imaging screens, we reveal that human Rab GTPases are common autophagy substrates. Importantly, we confirm the conservation of Rab GTPase autophagic degradation in different model organisms. Rab GTPases translocate to damaged mitochondria, lipid droplets, and invading Salmonella-containing vacuoles (SCVs) to serve as degradation signals. Furthermore, they facilitate mitophagy, lipophagy, and xenophagy, respectively, by recruiting receptors. This interplay between Rab GTPases and receptors may ensure the de novo synthesis of isolation membranes around Rab-GTPase-labeled cargo, thereby mediating selective autophagy. These processes are further influenced by upstream regulators such as LRRK2, GDIs, and RabGGTase. In conclusion, this study unveils a conserved mechanism involving Rab GTPases as autophagy cues signals and proposes a model for the spatiotemporal control of selective autophagy.
Collapse
Affiliation(s)
- Pengwei Zhao
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Rui Tian
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Dandan Song
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Qi Zhu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Xianming Ding
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jianqin Zhang
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Beibei Cao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengyuan Zhang
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yilu Xu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Jie Fang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jieqiong Tan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Cong Yi
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongguang Xia
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Center for Metabolism Research, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, China
| |
Collapse
|
6
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
7
|
Xu J, Ren F, Wang J, Liu J, Cui X, Hao J, Yang W, Zhang Y, Cao D, Li L, Wang H. Tubeimoside I induces mitophagy by activating the PINK1/Parkin/Mfn2 signaling pathway in acute myeloid leukemia cells. Transl Oncol 2025; 55:102355. [PMID: 40112502 PMCID: PMC11979407 DOI: 10.1016/j.tranon.2025.102355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Acute myeloid leukemia (AML) is the most prevalent kind of acute leukemia in adults. Despite the availability of new targeted therapies, AML remains connected with a poor prognosis and decreased rate of survival. Tubeimoside I (TBMS1), a critical compound extracted from Bolbostemma paniculatum, has demonstrated potential anticancer effects in lung and colorectal cancers. Nevertheless, the TBMS1 anticancer pathway against AML is still elusive. This study aimed to explore the potential role of TBMS1 in anti-AML and its molecular mechanism. In vitro, TBMS1 treatment suppressed AML cells proliferation, induced apoptosis, and mitochondrial damage, and elevated ROS levels. Network pharmacological analysis suggested, and subsequent studies confirmed, that TBMS1 induced mitophagy in AML cells by modulating the PINK1/Parkin/Mfnh2 signaling pathway, an effect that was effectively reversed following PINK1 knockdown. In vivo, TBMS1 treatment suppressed the proliferation of AML cells after 21 days, improved the survival rates of nude mice, and showed no evident organ toxicity. These evidences suggest that TBMS1 may have significant therapeutic potential in treating AML.
Collapse
Affiliation(s)
- Jing Xu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan 030001, China
| | - Fanggang Ren
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Jinjuan Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Jianbing Liu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaohua Cui
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Jianqing Hao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Wanfang Yang
- School of Basic Medicine, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Yaofang Zhang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Dongmin Cao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Zhongshan 528437, China.
| | - Li Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan 030001, China.
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China; School of Basic Medicine, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| |
Collapse
|
8
|
Yan W, Xiang S, Feng J, Zu X. Role of ubiquitin-specific proteases in programmed cell death of breast cancer cells. Genes Dis 2025; 12:101341. [PMID: 40083330 PMCID: PMC11904532 DOI: 10.1016/j.gendis.2024.101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related deaths among women worldwide. Great progress has been recently achieved in controlling breast cancer; however, mortality from breast cancer remains a substantial challenge, and new treatment mechanisms are being actively sought. Programmed cell death (PCD) is associated with the progression and treatment of many types of human cancers. PCD can be divided into multiple pathways including autophagy, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis, and anoikis. Ubiquitination is a post-translational modification process in which ubiquitin, a 76-amino acid protein, is coupled to the lysine residues of other proteins. Ubiquitination is involved in many physiological events and promotes cancer development and progression. This review elaborates the role of ubiquitin-specific protease (USP) in programmed cell death, which is common in breast cancer cells, and lays the foundation for tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
| | | | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
9
|
Kaur V, Sunkaria A. Unlocking the therapeutic promise of miRNAs in promoting amyloid-β clearance for Alzheimer's disease. Behav Brain Res 2025; 484:115505. [PMID: 40010509 DOI: 10.1016/j.bbr.2025.115505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/06/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
Alzheimer's disease (AD) is a neurological disorder that affects cognition and behavior, accounting for 60-70 % of dementia cases. Its mechanisms involve amyloid aggregates, hyperphosphorylated tau tangles, and loss of neural connections. Current treatments have limited efficacy due to a lack of specific targets. Recently, microRNAs (miRNAs) have emerged as key modulators in AD, regulating gene expression through interactions with mRNA. Dysregulation of specific miRNAs contributes to disease progression by disrupting clearance pathways. Antisense oligonucleotide (ASO)-based therapies show promise for AD treatment, particularly when combined with miRNA mimics or antagonists, targeting complex regulatory networks. However, miRNAs can interact with each other, complicating cellular processes and potentially leading to side effects. Our review emphasizes the role of miRNAs in regulating amyloid-beta (Aβ) clearance and highlights their potential as therapeutic targets and early biomarkers for AD, underscoring the need for further research to enhance their efficacy and safety.
Collapse
Affiliation(s)
- Vajinder Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India.
| |
Collapse
|
10
|
Long Y, Li Y, Ma Z, Xie Y, Zhao H, Zhang M, Liu R. Epimedii Folium and Ligustri Lucidi Fructus synergistically delay renal aging through AMPK/ULK1/Bcl2L13-mediated mitophagy. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119668. [PMID: 40122318 DOI: 10.1016/j.jep.2025.119668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of aging is attributed to kidney essence insufficiency and gradual loss of kidney function. The combination of Epimedii Folium and Ligustri Lucidi Fructus (ELL) is traditionally recognized to tonify kidney yin and yang and has significant efficacy in delaying aging and aging-related diseases, but little is known about the exact mechanism. AIM OF THE STUDY The research focuses on the mechanism of delaying renal aging by which ELL regulates mitophagy through serine/threonine kinase AMP-activated protein kinase (AMPK)/UNC-51- like autophagy activating kinase 1 (ULK1)/B-cell lymphoma-2-like protein 13 (Bcl2L13) in vivo. MATERIALS AND METHODS We employed a rat model of natural aging, using rats of different ages as dynamic controls, and a natural aging mouse model to evaluate the effects of ELL on delaying renal aging via AMPK/ULK1/Bcl2L13. The assessment included renal histopathology, oxidative stress, cell senescence, mitochondrial dynamics, mitophagy, and the AMPK/ULK1/Bcl2L13 signaling pathway. In the aging rat model, network pharmacology and proteomics were combined to dissect the renal aging process, and a Multilayer Perceptron (MLP) -artificial neural networks (ANN) model was used to evaluate the effects of ELL comprehensively. In the aging mouse model, the AMPK inhibitor dorsomorphin was applied to assess whether the AMPK signaling pathway was involved in ELL-induced mitophagy. RESULTS Compared with the young rats, the kidney exhibited signs of degenerative pathologies and increased oxidative stress in 17-month-old rats. A thorough analysis identified the mitochondrial protein Bcl2L13 as a crucial biomarker associated with renal aging. The AMPK/ULK1/Bcl2L13 pathway significantly regulated mitochondrial function and mitophagy, which were potential mechanisms underlying renal aging. In contrast to aged rats, the renal pathological changes and cell senescence in rats treated with ELL were significantly mitigated, the antioxidant capacity, mitochondrial dynamics, and mitophagy were improved, and the expression of AMPK/ULK1/Bcl2L13 was upregulated. After the application of AMPK inhibitor dorsomorphin, the effects of ELL were reversed. It appears that ELL modulates the AMPK/ULK1/Bcl2L13 signaling pathway, and upregulates mitophagy to potentially decelerate renal aging. CONCLUSIONS The findings indicate that aging kidneys display mitochondrial dysfunction, disorganization of mitochondria, and a decrease in mitophagy. Concurrently, ELL significantly regulates mitochondrial dynamics and mitophagy via AMPK/ULK1/Bcl2L13. This regulation helps mitigate mitochondrial dysfunction, suggesting ELL as a promising herbal remedy for delaying renal aging and age-related kidney diseases.
Collapse
Affiliation(s)
- Yuting Long
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Yuman Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Zaina Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Yonghao Xie
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of Traditional Chinese Medicine Collateral Disease Theory Research, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing, 100069, China
| | - Minyu Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of Traditional Chinese Medicine Collateral Disease Theory Research, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing, 100069, China.
| | - Renhui Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of Traditional Chinese Medicine Collateral Disease Theory Research, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing, 100069, China.
| |
Collapse
|
11
|
Li Z, Yi H, Li Y, Yang J, Guo P, Han F. Identification and validation of a novel autophagy-related biomarker in obstructive sleep apnea syndrome. Sleep 2025; 48:zsae287. [PMID: 39665515 DOI: 10.1093/sleep/zsae287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea syndrome is closely associated with tumor growth. Chronic intermittent hypoxia promotes autophagy and is related to malignant tumor development. However, the role of autophagy in obstructive sleep apnea syndrome progression remains unclear. METHODS obstructive sleep apnea syndrome datasets (GSE135917 and GSE38792) from Gene Expression Omnibus were analyzed to identify differentially expressed genes and autophagy-related differentially expressed genes. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analysis were conducted, and a protein-protein interaction network identified hub genes. Colorectal cancer datasets from The Cancer Genome Atlas were used for differential expression and survival analyses, along with gene set enrichment analysis and immune infiltration analysis. Chronic intermittent hypoxia-induced autophagy and oxidative stress were investigated in Sprague-Dawley rats using reactive oxygen species assays. Hub genes were validated in rats and obstructive sleep apnea syndrome patient samples. RESULTS Gene set enrichment analysis revealed significant differences in autophagy-related gene expression among obstructive sleep apnea syndrome patients. Hub genes ATG5, CASP1, MAPK8, EIF4G1, and TANK-binding kinase 1 were identified, with ATG5 and TANK-binding kinase 1 validated. Autophagy-related differentially expressed genes were predominantly upregulated in colorectal cancer tissues. TANK-binding kinase 1 expression in colorectal cancer patients was associated with enhanced sensitivity to immunotherapy and CD8 + T cell, macrophage, and regulatory T cell infiltration, potentially influencing the immune microenvironment. The animal experiments showed that chronic intermittent hypoxia increased reactive oxygen species levels, suggesting that chronic intermittent hypoxia plays a role in autophagy. TANK-binding kinase 1 expression was significantly higher in obstructive sleep apnea syndrome patients than in controls, and continuous positive airway pressure did not alter TANK-binding kinase 1 levels. CONCLUSIONS This study is the first to describe the potential contribution of TANK-binding kinase 1 to the development of obstructive sleep apnea syndrome and its potential as a novel biomarker and potential therapeutic target for obstructive sleep apnea syndrome.
Collapse
Affiliation(s)
- Zhiyong Li
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Huijie Yi
- Department of Sleep Medicine, Peking University People's Hospital, Xicheng, Beijing, China
| | - Yuxi Li
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Jie Yang
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Peng Guo
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Fang Han
- Department of Sleep Medicine, Peking University People's Hospital, Xicheng, Beijing, China
| |
Collapse
|
12
|
Sampaio Cruz M, Manso AM, Soto-Hermida A, Bushway P, Silver E, Gunes BB, Tang Z, Gonzalez G, Lau S, Arbayo J, Najor RH, Chi L, Gu Y, Feng W, Cowling RT, Gustafsson AB, Chen J, Adler ED. Overlapping functions between Lamp2a and Lamp2b in cardiac autophagy. Autophagy 2025:1-12. [PMID: 40202173 DOI: 10.1080/15548627.2025.2484620] [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: 12/27/2023] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/10/2025] Open
Abstract
LAMP2 is a ubiquitously expressed protein critical for autophagy. Alternative splicing gives rise to three isoforms. However, the roles of major LAMP2 isoforms in the heart are not known. To address this knowledge gap, we generated lamp2a and lamp2b knockout (KO) mice to investigate the role of these isoforms in heart function and autophagy. Deletion of either Lamp2a or Lamp2b did not alter cardiac structure or function. Lack of all LAMP2 isoforms led to increased cardiac fibrosis and reduced survival during pressure overload, which were not observed in lamp2a or lamp2b KO mice. Also, LAMP2B loss did not affect levels of the autophagy markers LC3-II and SQSTM1/p62. Conversely, LAMP2A was upregulated in hearts lacking LAMP2B, potentially preserving autophagy and cardiac function. Reintroducing LAMP2A in lamp2 KO mice effectively reduced autophagosome accumulation and improved cardiac function. Overall, these data support LAMP2 isoform functional redundancy in the myocardium under pathological conditions.Abbreviations: AAV: adeno-associated virus; ACTA2: actin alpha 2, smooth muscle, aorta; CMA: chaperone-mediated autophagy; KO: knockout; LAMP2: lysosomal-associated membrane protein 2; LV: Left ventricle; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; NPPA: natriuretic peptide type A; NPPB: natriuretic peptide type B; SQSTM1/p62: sequestosome 1; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; TAC: transverse aortic constriction; WT: wild type.
Collapse
Affiliation(s)
- Marina Sampaio Cruz
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Ana Maria Manso
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Angel Soto-Hermida
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Paul Bushway
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Elizabeth Silver
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Betul Beyza Gunes
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Zhiyuan Tang
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Giovanni Gonzalez
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Sharon Lau
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Jordan Arbayo
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Liguo Chi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Yusu Gu
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Wei Feng
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Randy T Cowling
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Asa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Ju Chen
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| | - Eric D Adler
- Department of Medicine, Division of Cardiology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
13
|
Zou Y, Yang L, Zhu J, Fan J, Zheng H, Liao X, Yang Z, Zhang K, Jia H, Konnerth A, Wang YJ, Zhang C, Zhang Y, Li SC, Chen X. Pitolisant alleviates brain network dysfunction and cognitive deficits in a mouse model of Alzheimer's disease. Transl Psychiatry 2025; 15:126. [PMID: 40185739 PMCID: PMC11971262 DOI: 10.1038/s41398-025-03358-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Histamine H3 receptor (H3R) antagonists regulate histamine release that modulates neuronal activity and cognitive function. Although H3R is elevated in Alzheimer's disease (AD) patients, whether H3R antagonists can rescue AD-associated neural impairments and cognitive deficits remains unknown. Pitolisant is a clinically approved H3R antagonist/inverse agonist that treats narcolepsy. Here, we find that pitolisant reverses AD-like pathophysiology and cognitive impairments in an AD mouse model. Behavioral assays and in vivo wide-field Ca2+ imaging revealed that recognition memory, learning flexibility, and slow-wave impairment were all improved following the 15-day pitolisant treatment. Improved recognition memory was tightly correlated with slow-wave coherence, suggesting slow waves serve as a biomarker for treatment response and for AD drug screening. Furthermore, pitolisant reduced amyloid-β deposition and dystrophic neurites surrounding plaques, and enhanced neuronal lysosomal activity, inhibiting which blocked cognitive and slow-wave restoration. Our findings identify pitolisant as a potential therapeutic agent for AD treatments.
Collapse
Affiliation(s)
- Yang Zou
- Guangxi Key Laboratory of Special Biomedicine/Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
| | - Linhan Yang
- Guangxi Key Laboratory of Special Biomedicine/Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
| | - Jiahui Zhu
- Guangxi Key Laboratory of Special Biomedicine/Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
| | - Jihua Fan
- Guangxi Key Laboratory of Special Biomedicine/Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
| | - Hanrun Zheng
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, 400038, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, 400038, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, 400038, China
- LFC Laboratory and Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China
| | - Hongbo Jia
- Guangxi Key Laboratory of Special Biomedicine/Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
- Institute of Neuroscience and Munich Cluster for Systems Neurology, Technical University Munich, 80802, Munich, Germany
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Arthur Konnerth
- Institute of Neuroscience and Munich Cluster for Systems Neurology, Technical University Munich, 80802, Munich, Germany
| | - Yan-Jiang Wang
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, 400038, China
- LFC Laboratory and Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China
| | - Chunqing Zhang
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, 400038, China.
- LFC Laboratory and Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, China.
| | - Sunny C Li
- LFC Laboratory and Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
- NewLight Neuroscience Unit, Chongqing, 400064, China.
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, 400038, China.
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing, 400038, China.
- LFC Laboratory and Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| |
Collapse
|
14
|
Qu H, Yuan X, Huang K, Liu D. AKT/mTOR mediated autophagy contributes to the self-replication of canine influenza virus in vivo and in vitro. Cell Signal 2025; 128:111648. [PMID: 39929352 DOI: 10.1016/j.cellsig.2025.111648] [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: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The prevalence and spread of canine influenza virus (CIV) pose a threat to the health of dogs and humans. Some studies have shown that autophagy is closely related to virus replication, but the exact relationship between CIV replication and autophagy is still unclear. Therefore, this study investigated the effects of autophagy on CIV replication in vitro and in vivo. The data showed that CIV infection significantly caused respiratory tract damage in mice, upregulated the mRNA/protein levels of CIV replication-related genes and autophagy-related genes. In addition, the activation of autophagy by rapamycin (Rapa) significantly intensified the CIV replication and the respiratory tract damage of mice, while the inhibition of autophagy by 3-Methyladenine (3-MA) significantly alleviated these effects. Data of MDCK cells also demonstrated that CIV promoted self-replication through activating autophagy, and the upregulation of AKT/mTOR by insulin significantly inhibited the CIV replication. In summary, this study showed that CIV could promote self-replication by activating AKT/mTOR mediated autophagy, which provides new ideas for the prevention and treatment of canine influenza.
Collapse
Affiliation(s)
- Haobo Qu
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, People's Republic of China
| | - Xin Yuan
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, People's Republic of China
| | - Kehe Huang
- College of Veterinary Medicine, Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing, Jiangsu Province 210095, People's Republic of China
| | - Dandan Liu
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou 310058, People's Republic of China.
| |
Collapse
|
15
|
Li W, Dong W, Zhu Z, Cao B, Xu T, Sun Y. pacsin1 inhibits antiviral immunity by promoting MITA degradation through autophagy in miiuy croaker, Miichthysmiiuy. FISH & SHELLFISH IMMUNOLOGY 2025; 159:110182. [PMID: 39923887 DOI: 10.1016/j.fsi.2025.110182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
Pacsin1 is a crucial protein involved in vesicle formation and transport, and its role in neuronal development and cytosolic dynamics has been extensively studied. However, its involvement in immune regulation still needs to be better understood. In this study, we show that pacsin1 exerts a negative regulatory effect on RLR-mediated signaling pathways activated by SCRV or poly(I:C), thereby inhibiting MITA-mediated antiviral responses. Mechanistically, pacsin1 facilitates the degradation of MITA, thus impeding immune signaling. Additionally, overexpression of pacsin1 promotes the conversion of LC3B-I to LC3B-II, while treatment with the autophagy inhibitor ammonium chloride results in the accumulation of LC3B-II and prevents pacsin1-mediated MITA degradation. Our findings suggest that pacsin1 targets MITA for autophagic degradation, thereby suppressing the innate antiviral response in fish.
Collapse
Affiliation(s)
- Wenxin Li
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Wenjing Dong
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Zhihuang Zhu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Baolan Cao
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China.
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, China.
| |
Collapse
|
16
|
Chen Y, Wang Z, Ma Q, Sun C. The role of autophagy in fibrosis: Mechanisms, progression and therapeutic potential (Review). Int J Mol Med 2025; 55:61. [PMID: 39950330 PMCID: PMC11878481 DOI: 10.3892/ijmm.2025.5502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Various forms of tissue damage can lead to fibrosis, an abnormal reparative reaction. In the industrialized countries, 45% of deaths are attributable to fibrotic disorders. Autophagy is a highly preserved process. Lysosomes break down organelles and cytoplasmic components during autophagy. The cytoplasm is cleared of pathogens and dysfunctional organelles, and its constituent components are recycled. With the growing body of research on autophagy, it is becoming clear that autophagy and its associated mechanisms may have a role in the development of numerous fibrotic disorders. However, a comprehensive understanding of autophagy in fibrosis is still lacking and the progression of fibrotic disease has not yet been thoroughly investigated in relation to autophagy‑associated processes. The present review focused on the latest findings and most comprehensive understanding of macrophage autophagy, endoplasmic reticulum stress‑mediated autophagy and autophagy‑mediated endothelial‑to‑mesenchymal transition in the initiation, progression and treatment of fibrosis. The article also discusses treatment strategies for fibrotic diseases and highlights recent developments in autophagy‑targeted therapies.
Collapse
Affiliation(s)
| | | | - Qinghong Ma
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Chao Sun
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| |
Collapse
|
17
|
Marino Y, Inferrera F, Genovese T, Cuzzocrea S, Fusco R, Di Paola R. Mitochondrial dynamics: Molecular mechanism and implications in endometriosis. Biochimie 2025; 231:163-175. [PMID: 39884375 DOI: 10.1016/j.biochi.2025.01.012] [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: 12/21/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Endometriosis affects about 10 % of women of reproductive age, leading to a disabling gynecologic condition. Chronic pain, inflammation, and oxidative stress have been identified as the molecular pathways involved in the progression of this disease, although its precise etiology remains uncertain. Although mitochondria are considered crucial organelles for cellular activity, their dysfunction has been linked to the development of this disease. The purpose of this review is to examine the functioning of the mitochondrion in endometriosis: in particular, we focused on the mitochondrial dynamics of biogenesis, fusion, and fission. Since excessive mitochondrial activity is reported to affect cell proliferation, we also considered mitophagy as a mechanism involved in limiting disease development. To better understand mitochondrial activity, we also considered alterations in circadian rhythms, the gut microbiome, and estrogen receptors: indeed, these mechanisms are also involved in the development of endometriosis. In addition, we focused on recent research about the impact of numerous substances on mitochondrial activity; some of them may offer a future breakthrough in endometriosis treatment by acting on mitochondria and inhibiting cell proliferation.
Collapse
Affiliation(s)
- Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166, Messina, Italy.
| | - Francesca Inferrera
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166, Messina, Italy.
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166, Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166, Messina, Italy; Link Campus University, Via del Casale di San Pio V, 44, Italy.
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166, Messina, Italy.
| | - Rosanna Di Paola
- Department of Veterinary Sciences, 98168, University of Messina, Messina, Italy.
| |
Collapse
|
18
|
Wang S, Ren J, Chi J, Guan Y, Zheng R, Wang J, Liu X, Huang H. RHOD mediates ATG9A trafficking to promote autophagosome formation during autophagy in cancer. Autophagy 2025:1-19. [PMID: 40143438 DOI: 10.1080/15548627.2025.2484604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 03/28/2025] Open
Abstract
ATG9A is a transmembrane protein essential for macroautophagy/autophagy that drives autophagosome formation by delivering essential proteins and lipids to the phagophore through vesicle trafficking. Here, we demonstrate that the atypical Rho GTPase RHOD is required for ATG9A trafficking and stimulates autophagosome formation. Upon starvation, RHOD interacted with ATG9A and accompanied ATG9A trafficking from the Golgi toward phagophores. In addition, starvation-induced high levels of RHOD resulted in Golgi fragmentation to further promote ATG9A vesicle export from the trans-Golgi network to the peripheral region. Loss of RHOD suppressed ATG9A trafficking and reduced the distribution of ATG9A on the phagophore. Moreover, WHAMM (WASP homolog associated with actin, golgi membranes and microtubules) forms a complex with RHOD and participates in this process in a RHOD-dependent manner. Importantly, RHOD mutants, which lack the exon II-containing effector region motif that is required for ATG9A binding or lack the CAAX box that is responsible for membrane targeting, fail to stimulate ATG9A trafficking and autophagosome formation. Furthermore, RHOD plays a distinct suppressor role in tumor development, partly associated with its regulatory effect on autophagy. These findings reveal the important roles of RHOD in autophagy and tumor development.Abbreviation: ATG9A: autophagy related 9A; BafA1: bafilomycin A1; BiFC: bimolecular fluorescence complementation; co-IP: co-immunoprecipitation; EBSS: Earle's balanced salt solution; FM: full culture medium; KO: knockout; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PUP-IT: pupylation-based interaction tagging; RHOD: ras homolog family member D; SQSTM1: sequestosome 1; TGN: trans-Golgi network; VC: Venus C-terminal; VN: Venus N-terminal; WHAMM: WASP homolog associated with actin, golgi membranes and microtubules; WIPI2: WD repeat domain, phosphoinositide interacting 2; WT: wild-type; 3-MA: phosphatidylinositol 3-kinase (PtdIns3K) inhibitor 3-methyladenine.
Collapse
Affiliation(s)
- Sijia Wang
- Beijing Key Laboratory of Environmental and Viral Oncology, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jing Ren
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jinghan Chi
- Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Yifei Guan
- Beijing Key Laboratory of Environmental and Viral Oncology, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Ran Zheng
- Beijing Key Laboratory of Environmental and Viral Oncology, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Juan Wang
- Beijing Key Laboratory of Environmental and Viral Oncology, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xinhui Liu
- Beijing Key Laboratory of Environmental and Viral Oncology, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Hua Huang
- Beijing Key Laboratory of Environmental and Viral Oncology, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
19
|
Dabravolski SA, Kalmykov VA, Maksaeva AO, Rozhkova UV, Lapshina KO, Orekhov AN. Necroptosis in myocardial ischaemia-reperfusion injury: current update on mechanisms, therapeutic targets, and translational potential. Apoptosis 2025:10.1007/s10495-025-02108-x. [PMID: 40146485 DOI: 10.1007/s10495-025-02108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2025] [Indexed: 03/28/2025]
Abstract
Necroptosis is a programmed form of cell death that has gained significant attention in the field of cardiovascular research due to its involvement in myocardial infarction (MI) and myocardial ischaemia-reperfusion (I/R) injury. Unlike apoptosis, necroptosis elicits a pro-inflammatory response, contributing to myocardial injury, fibrosis, and adverse remodelling. This review aims to provide an overview of the molecular mechanisms underlying necroptosis, with a particular focus on its role in myocardial I/R injury. Key regulatory proteins such as Receptor-interacting protein kinase 3 (RIPK3) and Mixed lineage kinase domain-like protein (MLKL) are central to the necroptotic process, mediating cell death and inflammation. The review discusses the potential of targeting necroptosis as a therapeutic strategy for managing cardiovascular diseases, particularly post-MI. The RIPK3-CaMKII-mitochondrial permeability transition pore (mPTP) pathway is identified as a critical signalling axis in necroptosis and its inhibition may offer protective benefits in myocardial injury. The review also considers the role of natural and chemical inhibitors and other genes in necroptosis regulation. Overall, targeting necroptosis represents a promising avenue for therapeutic intervention to mitigate cardiac injury, promote recovery, and improve long-term patient outcomes in cardiovascular diseases.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, 2161002, Karmiel, Israel.
| | - Vladislav A Kalmykov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Anastasia O Maksaeva
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
- Sechenov First Moscow State Medical University, 8, Trubetskaya Street, Building 2, Moscow, Russia, 119991
| | - Ulyana V Rozhkova
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, Russia, 125315
| | - Ksenia O Lapshina
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, Moscow, Russia, 117418
| | - Alexander N Orekhov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, Moscow, Russia, 117418
| |
Collapse
|
20
|
Yamamoto T. Autophagic stagnation: a key mechanism in kidney disease progression linked to aging and obesity. Clin Exp Nephrol 2025:10.1007/s10157-025-02653-4. [PMID: 40131605 DOI: 10.1007/s10157-025-02653-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Autophagy, a critical intracellular degradation and recycling pathway mediated by lysosomes, is essential for maintaining cellular homeostasis through the quality control of proteins and organelles. Our research focused on the role of proximal tubular autophagy in the pathophysiology of aging, obesity, and diabetes. Using a novel method to monitor autophagic flux in kidney tissue, we revealed that age-associated high basal autophagy supports mitochondrial quality control and delays kidney aging. However, an impaired ability to upregulate autophagy under additional stress accelerates kidney aging. In obesity induced by a high-fat diet, lysosomal dysfunction disrupts autophagy, leading to renal lipotoxicity. Although autophagy is initially activated to repair organelle membranes and maintain proximal tubular cell integrity, this demand overwhelms lysosomes, resulting in "autophagic stagnation" characterized by phospholipid accumulation. Similar lysosomal phospholipid accumulation was observed in renal biopsies from elderly and obese patients. We identified TFEB-mediated lysosomal exocytosis as a mechanism to alleviate lipotoxicity by expelling accumulated phospholipids. Therapeutically, interventions such as the SGLT2 inhibitor empagliflozin and eicosapentaenoic acid restore lysosomal function and autophagic activity. Based on these findings, we propose a novel disease concept, "Obesity-Related Proximal Tubulopathy." This study underscores autophagic stagnation as a key driver of kidney disease progression in aging and obesity, offering insights into the pathophysiology of kidney diseases and providing a foundation for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Box D11, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
21
|
Zhang R, Huang W, Zhao T, Fang J, Chang C, He D, Wang X. Comprehensive analysis and validation of autophagy-related gene in rheumatoid arthritis. Front Cell Dev Biol 2025; 13:1563911. [PMID: 40181826 PMCID: PMC11965638 DOI: 10.3389/fcell.2025.1563911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease in which autophagy is pivotal in its pathogenesis. This study aims to identify autophagy-related genes associated with RA and investigate their functional roles. Methods We performed mRNA sequencing to identify differentially expressed genes (DEGs) between RA and osteoarthritis (OA) and intersected these with autophagy-related genes to obtain autophagy-related DEGs (ARDEGs) in RA. Bioinformatics and machine learning approaches were used to identify key biomarkers. Functional experiments, including real-time cellular analysis (RTCA), scratch healing, and flow cytometry, were conducted to examine the effects of gene silencing on the proliferation and migration of MH7A cells. Results A total of 37 ARDEGs were identified in RA. Through bioinformatics analysis, interferon regulatory factor 4 (IRF4) emerged as a key hub gene, with its high expression confirmed in RA synovial tissues and RA FLS cells. IRF4 knockdown inhibited the proliferation and migration and promoted the death of MH7A cells. Conclusion IRF4 is an autophagy-related diagnostic biomarker for RA. Targeting IRF4 could serve as a potential diagnostic and therapeutic strategy for RA, although further clinical studies are required to validate its effectiveness.
Collapse
Affiliation(s)
- Runrun Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenhan Huang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jintao Fang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xinchang Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
22
|
Li M, Wei CB, Li HF, He K, Bai RJ, Zhang FJ. Osteopontin inhibits autophagy via CD44 and avβ3 integrin and promotes cell proliferation in osteoarthritic fibroblast-like synoviocytes. BMC Musculoskelet Disord 2025; 26:274. [PMID: 40102843 PMCID: PMC11916941 DOI: 10.1186/s12891-025-08509-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is closely related to aging, and autophagy is implicated in the retardation of aging. Activated synoviocytes play important roles in OA; the synoviocytes could produce osteopontin (OPN) and its main receptors CD44 and integrin, which are all involved in OA. The purpose of this study is to investigate whether OPN has an effect on autophagy in osteoarthritic synoviocytes. METHODS We cultured human OA fibroblast-like synoviocytes (FLS) and treated them with rhOPN and antibodies against CD44 and CD51/61 (αvβ3 integrin) or isotype IgG to block the interaction between receptors and ligands. Infection with lentivirus mRFP-GFP-LC3, laser confocal imaging and Western blotting were used to determine changes in the expression of autophagy markers, and cell proliferation of FLS was assessed with a CCK-8 assay. RESULTS Our results showed the expression level of autophagy marker protein LC3 II and the mRFP-GFP-LC3 puncta were significantly decreased after treatment with rhOPN when compared with the control group, when the FLS were incubated with antibodies against CD44 or CD51/61 (αvβ3 integrin) or with control isotype IgG for 1 h, followed by rhOPN treatment for 48 h, rhOPN could suppress the relative expression of LC3 II and Beclin1 via integrin and CD44 in the FLS, CCK-8 assay also showed that rhOPN significantly increased the cell proliferation and viability of FLS. CONCLUSIONS OPN could inhibit autophagy via CD44 and αvβ3 integrin and promote the proliferation of FLS, playing an important role in OA synovitis.
Collapse
Affiliation(s)
- Min Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Chang-Bao Wei
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Hai-Feng Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Rui-Jun Bai
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China.
| | - Fang-Jie Zhang
- Department of Emergency Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), No.87 Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
23
|
Hassan MAM, Fahmy MI, Azzam HN, Ebrahim YM, El-Shiekh RA, Aboulmagd YM. Multifaceted therapeutic potentials of catalpol, an iridoid glycoside: an updated comprehensive review. Inflammopharmacology 2025:10.1007/s10787-025-01694-1. [PMID: 40097877 DOI: 10.1007/s10787-025-01694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Catalpol, classified as an iridoid glucoside, is recognized for its significant role in medicine, particularly in the treatment of various conditions such as diabetes mellitus, neuronal disorders, and inflammatory diseases. This review aims to evaluate the biological implications of catalpol and the mechanisms underlying its diverse pharmacological effects. A thorough exploration of existing literature was conducted utilizing the keyword "Catalpol" across prominent public domains like Google Scholar, PubMed, and EKB. Catalpol has demonstrated a diverse array of pharmacological effects in experimental models, showcasing its anti-diabetic, cardiovascular-protective, neuroprotective, anticancer, hepatoprotective, anti-inflammatory, and antioxidant properties. In summary, catalpol manifests a spectrum of biological effects through a myriad of mechanisms, prominently featuring its anti-inflammatory and antioxidant capabilities. Its diverse pharmacological profile underscores its potential for therapeutic applications across a range of conditions. Further research is warranted to fully elucidate the clinical implications of catalpol and optimize its use in medical practice.
Collapse
Affiliation(s)
- Mennat-Allah M Hassan
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Hany N Azzam
- Pharmacy Practice Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Yara M Aboulmagd
- Department of Pharmacology & Toxicology Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
24
|
Smith HA, Templeman I, Davis M, Slater T, Clayton DJ, Varley I, James LJ, Middleton B, Johnston JD, Karagounis LG, Tsintzas K, Thompson D, Gonzalez JT, Walhin JP, Betts JA. Characterizing 24-Hour Skeletal Muscle Gene Expression Alongside Metabolic and Endocrine Responses Under Diurnal Conditions. J Clin Endocrinol Metab 2025; 110:e1017-e1030. [PMID: 38779872 PMCID: PMC11913097 DOI: 10.1210/clinem/dgae350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
CONTEXT Skeletal muscle plays a central role in the storage, synthesis, and breakdown of nutrients, yet little research has explored temporal responses of this human tissue, especially with concurrent measures of systemic biomarkers of metabolism. OBJECTIVE To characterize temporal profiles in skeletal muscle expression of genes involved in carbohydrate metabolism, lipid metabolism, circadian clocks, and autophagy and descriptively relate them to systemic metabolites and hormones during a controlled laboratory protocol. METHODS Ten healthy adults (9M/1F, [mean ± SD] age 30 ± 10 years; BMI 24.1 ± 2.7 kg·m-2) rested in the laboratory for 37 hours with all data collected during the final 24 hours (08:00-08:00 hours). Participants ingested hourly isocaloric liquid meal replacements alongside appetite assessments during waking before a sleep opportunity from 22:00 to 07:00 hours. Blood samples were collected hourly for endocrine and metabolite analyses, with muscle biopsies occurring every 4 hours from 12:00 to 08:00 hours the following day to quantify gene expression. RESULTS Plasma insulin displayed diurnal rhythmicity peaking at 18:04 hours. Expression of skeletal muscle genes involved in carbohydrate metabolism (Name, Acrophase [hours]: GLUT4, 14:40; PPARGC1A, 16:13; HK2, 18:24) and lipid metabolism (FABP3, 12:37; PDK4, 05:30; CPT1B, 12:58) displayed 24-hour rhythmicity that reflected the temporal rhythm of insulin. Equally, circulating glucose (00:19 hours), nonesterified fatty acids (04:56), glycerol (04:32), triglyceride (23:14), urea (00:46), C-terminal telopeptide (05:07), and cortisol (22:50) concentrations also all displayed diurnal rhythmicity. CONCLUSION Diurnal rhythms were present in human skeletal muscle gene expression as well systemic metabolites and hormones under controlled diurnal conditions. The temporal patterns of genes relating to carbohydrate and lipid metabolism alongside circulating insulin are consistent with diurnal rhythms being driven in part by the diurnal influence of cyclic feeding and fasting.
Collapse
Affiliation(s)
- Harry A Smith
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Iain Templeman
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Max Davis
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Tommy Slater
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK, NG1 4FQ
| | - David J Clayton
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK, NG1 4FQ
| | - Ian Varley
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK, NG1 4FQ
| | - Lewis J James
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK, LE11 3TU
| | - Benita Middleton
- Section of Chronobiology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK, GU2 7XH
| | - Jonathan D Johnston
- Section of Chronobiology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK, GU2 7XH
| | - Leonidas G Karagounis
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
- Mary MacKillop Institute for Health Research (MMIHR), Australian Catholic University (ACU), Melbourne, VIC 3000, Australia
| | - Kostas Tsintzas
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK, NG7 2UH
| | - Dylan Thompson
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Javier T Gonzalez
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - Jean-Philippe Walhin
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| | - James A Betts
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, UK, BA2 7AY
| |
Collapse
|
25
|
Saberiyan M, Gholami S, Ejlalidiz M, Rezaeian Manshadi M, Noorabadi P, Hamblin MR. The dual role of chaperone-mediated autophagy in the response and resistance to cancer immunotherapy. Crit Rev Oncol Hematol 2025; 210:104700. [PMID: 40086769 DOI: 10.1016/j.critrevonc.2025.104700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer immunotherapy has become a revolutionary strategy in oncology, utilizing the host immune system to fight malignancies. Notwithstanding major progress, obstacles such as immune evasion by tumors and the development of resistance still remain. This manuscript examines the function of chaperone-mediated autophagy (CMA) in cancer biology, focusing on its effects on tumor immunotherapy response and resistance. CMA is a selective degradation mechanism for cytosolic proteins, which is crucial for sustaining cellular homeostasis and regulating immune responses. By degrading specific proteins, CMA can either facilitate tumor progression in stressful conditions, or promote tumor suppression by removing oncogenic factors. This double-edged sword highlights the complexity of CMA in cancer progression and its possible effect on treatment results. Here we clarify the molecular mechanisms by which CMA can regulate the immune response and its possible role as a therapeutic target for improving the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Mohammadreza Saberiyan
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sarah Gholami
- Young Researchers and Ellie Club, Babol Branch. Islamic Azad University, Babol, Iran
| | - Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadsadegh Rezaeian Manshadi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Noorabadi
- Department of Internal Medicine, School of Medicine, Urmia University of Medical sciences, Urmia, Iran.
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa.
| |
Collapse
|
26
|
Wang Y, Dai Z, Xiao S, Zeng F, Lu Y. Profilin 1 induces drug resistance by downregulating CD138 expression via autophagy in multiple myeloma. Discov Oncol 2025; 16:284. [PMID: 40056333 DOI: 10.1007/s12672-025-02036-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 03/03/2025] [Indexed: 03/10/2025] Open
Abstract
Multiple myeloma (MM), a plasma cell-derived malignant hematological disease, is often treated with bortezomib, a highly effective first-generation proteasome inhibitor. However, resistance to bortezomib is a common occurrence. Profilin 1 (PFN1), a cytoskeleton-related gene known to promote autophagy in MM, induces this resistance to bortezomib, but it is unclear why. The aim of this study was to uncover the molecular mechanisms involved in bortezomib resistance, considering not only PFN1, but also CD138, a transmembrane proteoglycan that is a hallmark of plasma and MM cells. We detected CD138 and PFN1 in the bone marrow of patients with MM immunohistochemically. We also studied the Gene Expression Omnibus (GEO) data and found that CD138 was associated with PFN1 and autophagy. We then evaluated their expression in an MM cell line via western blot analysis, immunofluorescence assay, and flow cytometry; constructed PFN1-overexpressing and -knockdown cell lines; and detected ubiquitinated CD138 in the cells of both cell lines. Overexpression of PFN1 or PFN1-induced autophagy downregulated CD138 expression. Owing to the stemness and resistance of CD138- MM cells, inhibition of autophagy or CD138 overexpression reversed the resistance of PFN1-overexpressing MM cells to bortezomib, as indicated in our clonogenic, apoptosis, and CCK-8 assays. These results indicated that CD138 plays an important role in the resistance of MM cells to bortezomib. Targeting PFN1, CD138, or autophagic pathways may provide a promising therapeutic strategy for overcoming PFN1-induced drug resistance in MM.
Collapse
Affiliation(s)
- Ya Wang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - ZiYu Dai
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shengying Xiao
- Department of Oncology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), 89 Guhan Road, Furong, Changsha, 410002, China
| | - Furen Zeng
- Department of Oncology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), 89 Guhan Road, Furong, Changsha, 410002, China.
| | - YiChen Lu
- Department of Oncology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), 89 Guhan Road, Furong, Changsha, 410002, China.
| |
Collapse
|
27
|
Li T, Chen H, Guo Y, Huang M, Liu P, Aikemu A, Mohammadtursun N, Pan X, Yang X. Nuciferine Restores Autophagy via the PI3K-AKT-mTOR Pathway to Alleviate Renal Fibrosis in Diabetic Kidney Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5223-5235. [PMID: 39989251 DOI: 10.1021/acs.jafc.4c08844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Diabetic kidney disease (DKD) is one of the complications of diabetes mellitus, which triggers kidney fibrosis and eventually develops into end-stage renal disease. Nuciferine (NF) is one of the most important functional components in lotus leaves (LL), but its role and mechanism for the treatment of DKD are unclear. A high-fat-diet (HFD)-induced DKD model in KK-AY mice was established in this study. NF treatment significantly improved blood glucose and blood biochemical indices in DKD mice. Furthermore, NF reduced the levels of mALB, UCRE, Scr, and BUN in mice urine. Further, the extent of renal lesions in the mice in this study was at stage IV according to the Mogensen staging method. NF treatment was effective in ameliorating renal injury during this period. Concurrently, the protein levels of FN, N-cadherin, TGFβ, p-Smad3, p-PI3K, p-AKT, p-mTOR, and p62 were decreased. In contrast, the level of expression of Beclin-1 was increased. In the high glucose-exposed HK-2 cell model, the expression of p-PI3K, p-AKT, and p-mTOR was all downregulated, and autophagy proteins were increased after NF intervention. In addition, HK-2 cells were treated with high glucose in combination with Wortmannin and 3-MA, respectively. The results demonstrated that NF inhibited the expression of TGFβ and p-Smad3 by regulating autophagy through the PI3K-AKT-mTOR pathway, thereby ameliorating renal fibrosis at stage IV in mice. Therefore, LL can be used as a dietary component for the prevention of renal fibrosis in DKD patients.
Collapse
Affiliation(s)
- Tongqing Li
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Huijian Chen
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Yan Guo
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Mi Huang
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Pengxin Liu
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Ainiwaer Aikemu
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan 848000, China
| | - Nabijan Mohammadtursun
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan 848000, China
| | - Xin Pan
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Xinzhou Yang
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| |
Collapse
|
28
|
Ge G, Zhao W, Zhong Z, Huang Y, Hua Y, Chen K, Yu Y, Wu T, Lu Y, Yadav N, Zhang F. Acacetin ameliorates pressure overload-induced cardiac remodeling by targeting USP10 and inhibiting maladaptive cardiomyocyte autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156588. [PMID: 40118748 DOI: 10.1016/j.phymed.2025.156588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/11/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Numerous drugs have been developed to meet the critical demand for treatments inhibiting cardiac remodeling following cardiovascular disease. Acacetin is a flavonoid with potential therapeutic effects against various cardiovascular diseases. PURPOSE This study investigated the effect of acacetin on pressure overload-induced cardiac remodeling and its underlying molecular regulatory mechanisms. METHODS We simulated pressure overload-induced cardiac remodeling in male C57BL/6 mice by constricting the thoracic aortic arch and assessed the effect of acacetin on cardiac remodeling. RESULTS Acacetin significantly ameliorated cardiac remodeling by downregulating ubiquitin-specific peptidase 10 (USP10) protein expression and reducing autophagy levels in cardiomyocytes. These findings confirm that acacetin improves cardiac remodeling by suppressing cardiomyocyte autophagy and highlight the crucial role of USP10 in the Beclin 1 ubiquitination degradation-mediated inhibition of the cardiomyocyte autophagy signaling pathway. CONCLUSION These results suggest that acacetin is a promising candidate drug for the treatment of cardiac remodeling induced by pressure overload.
Collapse
Affiliation(s)
- Gaoyuan Ge
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China; Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Hanjiang Middle Road 368, Yangzhou 225000, Jiangsu, PR China
| | - Wei Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Zhuen Zhong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Youfu Huang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Yan Hua
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Kaiyan Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Yue Yu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Tianyu Wu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Yao Lu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Nishant Yadav
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Fengxiang Zhang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China.
| |
Collapse
|
29
|
Geurs S, Staessens E, Bredael K, Borghgraef S, De Ridder J, Persoons L, De Jonghe S, Schols D, Mann MK, Harding RJ, Franceus J, Desmet T, Van Hecke K, Clarisse D, De Bosscher K, D'hooghe M. Synthesis and functional screening of novel inhibitors targeting the HDAC6 zinc finger ubiquitin-binding domain. Eur J Med Chem 2025; 285:117208. [PMID: 39823806 DOI: 10.1016/j.ejmech.2024.117208] [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: 10/23/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025]
Abstract
Histone deacetylase 6 (HDAC6) is a promising target for treating neurodegenerative disorders, several cancer types and viral infections. Unique among HDACs, the HDAC6 isoform possesses a zinc finger ubiquitin-binding domain (UBD) crucial for managing misfolded protein aggregates and facilitating viral infection. HDAC6 binds aggregated polyubiquitinated proteins through its UBD, mediating their transport to the aggresome and subsequent removal via autophagy. Despite the importance of the UBD in proteostasis and viral infection, its pharmacological inhibition has been minimally explored thus far, with research largely focused on the deacetylase domain. We synthesized a diverse library of new compounds designed to target the HDAC6-UBD, termed HZUBi, with varied core structures including quinazolinone, oxindole and tetrahydrothiopyrano[4,3-b]indole, aimed at enhancing UBD interaction and extending into the side pocket. New structure-activity relationships were established, computational docking and molecular dynamics studies were performed and the functional impact of selected inhibitors was assessed in the context of multiple myeloma and viral infection. Several new HZUBi could displace a ubiquitin peptide from HDAC6-UBD in a differential manner, although to a lower extent than the literature reference compound HZUBi-3e. Despite exhibiting in vitro target engagement, neither HZUBi-3e nor its ester prodrug HZUBi-1e enhanced proteasome inhibitor-mediated multiple myeloma cell killing. Finally, none of the screened HZUBi triggered anti-viral activity.
Collapse
Affiliation(s)
- Silke Geurs
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium; Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Eleni Staessens
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Kato Bredael
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Stefaan Borghgraef
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Jordy De Ridder
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Leentje Persoons
- Molecular Genetics and Therapeutics in Virology and Oncology Research Group, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Steven De Jonghe
- Molecular Structural and Translational Virology Research Group, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Dominique Schols
- Molecular Structural and Translational Virology Research Group, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Mandeep K Mann
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Rachel J Harding
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jorick Franceus
- Center for Synthetic Biology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Tom Desmet
- Center for Synthetic Biology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kristof Van Hecke
- XStruct, Department of Chemistry, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Dorien Clarisse
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Karolien De Bosscher
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
30
|
Dražić Maras E, Kelam N, Racetin A, Haque E, Dražić M, Vukojević K, Katsuyama Y, Saraga-Babić M, Filipović N. Autophagy markers expression pattern in developing liver of the yotari (dab1 -/-) mice and humans. Acta Histochem 2025; 127:152224. [PMID: 39647211 DOI: 10.1016/j.acthis.2024.152224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Autophagy plays an important role in the physiology and pathology of the liver. Several negative autophagy regulators have been discovered, including epidermal growth factor receptor (EGFR), mediated by activation of the PI3K/Akt/mTOR signaling pathway. Disabled-1 (Dab1) is one of the mediating adaptor factors of PI3K/Akt/mTOR signaling pathways. We investigated the potential impact of Dab1 on autophagy-related markers (LC3B, LAMP2A, HSC70, and GRP78) in the developing liver by using a model of yotari mice and compared it with autophagy marker expression in human liver development. Mouse embryos were obtained at gestation days 13.5 and 15.5 (E13.5 and E15.5), and a total of 5 normal human conceptuses were obtained between gestation days 5 and 10. Histological sections were analyzed by immunohistochemistry. The highest expression of the early endosome-forming factor LC3B and the microautophagy factor LAMP2a was observed at the transition from embryonic to early fetal phase, whereas the expression of the chaperones HSC 70 and GRP78 was highest at embryonic phase. The expression patterns of three of these factors in mouse liver were different from those in human liver: the expression of LC3B was high at E13.5, that of HSC 70 at 15.5, whereas the expression of GRP78 did not change significantly. On the other hand, the expression pattern of LAMP2a was similar to that in human development and was higher at E15.5 than at E13.5. Moreover, knockout of Dab1 resulted in significantly lower expression of LC3B and LAMP2a in mouse embryo livers (at E13.5), indicating a possible role of Dab1 in regulating autophagy during embryonic development in the liver.
Collapse
Affiliation(s)
- Edita Dražić Maras
- Infectious Diseases Department, University Hospital of Split, Split 21000, Croatia
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split School of Medicine, Šoltanska 2A, Split 21000, Croatia
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split School of Medicine, Šoltanska 2A, Split 21000, Croatia
| | - Ejazul Haque
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split School of Medicine, Šoltanska 2A, Split 21000, Croatia
| | - Maja Dražić
- Department of Internal Medicine, Cardiology, General Hospital Knin, Knin 22300, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split School of Medicine, Šoltanska 2A, Split 21000, Croatia
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split School of Medicine, Šoltanska 2A, Split 21000, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split School of Medicine, Šoltanska 2A, Split 21000, Croatia.
| |
Collapse
|
31
|
Wang P, Li J, Li CG, Zhou X, Chen X, Zhu M, Wang H. Restoring Autophagy by Exercise Ameliorates Insulin Resistance Partly via Calcineurin-Driven TFEB Nuclear Translocation. Clin Exp Pharmacol Physiol 2025; 52:e70010. [PMID: 39787618 DOI: 10.1111/1440-1681.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 01/12/2025]
Abstract
Exercise activates autophagy and lysosome system in skeletal muscle, which are known to play an important role in metabolic adaptation. However, the mechanism of exercise-activated autophagy and lysosome system in obese insulin resistance remains covert. In this study, we investigated the role of exercise-induced activation of autophagy and lysosome system in improving glucose metabolism of skeletal muscle. Male C57BL/6 mice were randomly divided into five groups: the chow diet (CD) group, the high-fat diet (HFD) group, the high-fat diet plus exercise (HFD-E) group and the HFD-E treated with calcineurin inhibitor FK506 (HFD-E-F) or saline (HFD-E-S) groups. The mice in exercise groups (HFD-E, HFD-E-F and HFD-E-S) were subjected to aerobic treadmill exercise (speed at 12 m/min for 1 h per session, 0° slope, 5 days per week for 12 weeks). Mice of HFD-E-F group were intraperitoneally administered FK506 (1 mg/kg), once each day for 2 weeks before the end of exercise. Expressions pTFEB, T-TFEB and autophagy-lysosome markers, including Beclin1, LC3, ULK1, SQSTM1, LAMP1, CTSD and CTSL proteins in gastrocnemius muscle were analysed. We demonstrated that HFD induced insulin resistance and decreased autophagy-lysosomal proteins and the exercise significantly increased transcription factor EB (TFEB) translocation from the cytoplasm to the nucleus, restored the impaired autophagy-lysosomal-related protein expressions, and improved glucose metabolism. The increase in TFEB nuclear translocation was partly blocked by the calcineurin inhibitor FK506. Our results suggest that exercise promotes autophagy and lysosome restoration by regulating calcineurin-mediated TFEB nuclear translocation, ultimately alleviating HFD-induced insulin resistance in mice skeletal muscle.
Collapse
Affiliation(s)
- Ping Wang
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Jiaxin Li
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
| | - Xiaolong Chen
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Minghua Zhu
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Hongjiang Wang
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
32
|
Han X, Li P, Jiang M, Cao Y, Wang Y, Jiang L, Liu X, Wu W. Autophagy in skeletal muscle dysfunction of chronic obstructive pulmonary disease: implications, mechanisms, and perspectives. J Zhejiang Univ Sci B 2025; 26:227-239. [PMID: 40082202 PMCID: PMC11906388 DOI: 10.1631/jzus.b2300680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/22/2023] [Indexed: 03/16/2025]
Abstract
Skeletal muscle dysfunction is a common extrapulmonary comorbidity of chronic obstructive pulmonary disease (COPD) and is associated with decreased quality-of-life and survival in patients. The autophagy lysosome pathway is one of the proteolytic systems that significantly affect skeletal muscle structure and function. Intriguingly, both promoting and inhibiting autophagy have been observed to improve COPD skeletal muscle dysfunction, yet the mechanism is unclear. This paper first reviewed the effects of macroautophagy and mitophagy on the structure and function of skeletal muscle in COPD, and then explored the mechanism of autophagy mediating the dysfunction of skeletal muscle in COPD. The results showed that macroautophagy- and mitophagy-related proteins were significantly increased in COPD skeletal muscle. Promoting macroautophagy in COPD improves myogenesis and replication capacity of muscle satellite cells, while inhibiting macroautophagy in COPD myotubes increases their diameters. Mitophagy helps to maintain mitochondrial homeostasis by removing impaired mitochondria in COPD. Autophagy is a promising target for improving COPD skeletal muscle dysfunction, and further research should be conducted to elucidate the specific mechanisms by which autophagy mediates COPD skeletal muscle dysfunction, with the aim of enhancing our understanding in this field.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meiling Jiang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yuanyuan Cao
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Linhong Jiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China.
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai 201203, China.
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
33
|
Pabon A, Bhupana JN, Wong CO. Crosstalk between degradation and bioenergetics: how autophagy and endolysosomal processes regulate energy production. Neural Regen Res 2025; 20:671-681. [PMID: 38886933 PMCID: PMC11433889 DOI: 10.4103/nrr.nrr-d-23-02095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/08/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Cells undergo metabolic reprogramming to adapt to changes in nutrient availability, cellular activity, and transitions in cell states. The balance between glycolysis and mitochondrial respiration is crucial for energy production, and metabolic reprogramming stipulates a shift in such balance to optimize both bioenergetic efficiency and anabolic requirements. Failure in switching bioenergetic dependence can lead to maladaptation and pathogenesis. While cellular degradation is known to recycle precursor molecules for anabolism, its potential role in regulating energy production remains less explored. The bioenergetic switch between glycolysis and mitochondrial respiration involves transcription factors and organelle homeostasis, which are both regulated by the cellular degradation pathways. A growing body of studies has demonstrated that both stem cells and differentiated cells exhibit bioenergetic switch upon perturbations of autophagic activity or endolysosomal processes. Here, we highlighted the current understanding of the interplay between degradation processes, specifically autophagy and endolysosomes, transcription factors, endolysosomal signaling, and mitochondrial homeostasis in shaping cellular bioenergetics. This review aims to summarize the relationship between degradation processes and bioenergetics, providing a foundation for future research to unveil deeper mechanistic insights into bioenergetic regulation.
Collapse
Affiliation(s)
- Angelid Pabon
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | | | - Ching-On Wong
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| |
Collapse
|
34
|
Wen Y, Wang M, Liu X, Yin X, Gong S, Yin J. Deletion of FgAtg27 decreases the pathogenicity of Fusarium graminearum through influence autophagic process. Int J Biol Macromol 2025; 297:139818. [PMID: 39814284 DOI: 10.1016/j.ijbiomac.2025.139818] [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: 10/20/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Autophagy is a conserved and unique degradation system in eukaryotic cells, which plays crucial roles in the growth, development and pathogenesis of Fungi. Despite that, it is poorly understood in Fusarium graminearum currently. Here, we identified an autophagy gene FgAtg27 from F. graminearum, and investigated its possible roles in regulating morphogenesis and pathogenicity. Results showed that FgAtg27 is homologous to Saccharomyces cerevisiae Atg27 and with an active signal peptide at N-terminal. Then, the ΔFgAtg27 mutant was generated and gene deletion did not change growth and sporulation, whereas significantly decreased pathogenicity. FgAtg27 showed subcellular localization at pre-autophagosomal structure (PAS). After starvation induction, amount of autophagosomes in ΔFgAtg27 was significantly less than wild type and complemented strain, indicating that FgAtg27 deletion affects the autophagosome formation in F. graminearum. Meanwhile, under high Ca2+ concentration conditions, ΔFgAtg27 exhibited slowed growth, confirming that FgAtg27 also involved in F. graminearum's hyperosmotic reaction to Ca2+ concentration stress. In addition, yeast two-hybrid experiments, revealed that FgAtg27 interacts with the autophagy key protein FgAtg9. Collectively, we found that the deletion of FgAtg27 did not impact the growth phenotype of F. graminearum, whereas significantly reduced its pathogenicity and Ca2+ stress through affecting autophagic process.
Collapse
Affiliation(s)
- Yong Wen
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Mengru Wang
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Xi Liu
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Xiaohui Yin
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Shuangjun Gong
- Key Laboratory of Integrated Pest Management of Crops in Central China, Ministry of Agriculture, Hubei Key Laboratory of Crop Diseases, Insect Pests and Weeds Control, Institute of Plant Protection and Soil Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, Hubei, China
| | - Junliang Yin
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, MARA Key Laboratory of Sustainable Crop Production in the Middle Reaches of the Yangtze River (Co-construction by Ministry and Province), Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China.
| |
Collapse
|
35
|
Zeng L, Peng H, Sun L, Yu H, Wu Y. Exploring the mechanism of tiRNA-Val-CAC-002 in the pathogenesis of oral submucous fibrosis. J Oral Biosci 2025; 67:100588. [PMID: 39571899 DOI: 10.1016/j.job.2024.100588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 03/18/2025]
Abstract
OBJECTIVES Oral submucous fibrosis (OSF) is a chronic, progressive, and potentially malignant disease of the oral cavity. A previous study by our team found that the aberrant expression of tRNA-derived small RNA (tsRNA) was involved in the development of OSF, with tiRNA-Val-CAC-002 showing the most significant difference. This study aimed to investigate the effect of tiRNA-Val-CAC-002 on fibroblast activation and its underlying mechanisms, elucidate the pathogenesis of OSF, and explore new effective targets for OSF prevention and treatment. METHODS RT-PCR was used to detect tiRNA-Val-CAC-002 expression in OSF and arecoline-treated fibroblasts. Western blotting, MDC staining, and transmission electron microscopy validated the effects of arecoline and 002 on fibroblast autophagy. Western blotting was used to explore the signaling pathways related to tiRNA-Val-CAC-002 in OSF. RESULTS Arecoline promotes fibroblast (FB) activation by upregulating tiRNA-Val-CAC-002. Arecoline stimulation and tiRNA-Val-CAC-002 overexpression activated fibroblasts by promoting autophagy. tiRNA-Val-CAC-002 regulates PI3K/AKT by mediating ITGB3 expression. CONCLUSIONS Arecoline upregulates tiRNA-Val-CAC-002 expression in fibroblasts. Moreover, tiRNA-Val-CAC-002 may activate the autophagy of fibroblasts in OSF by ITGB3/PI3K/AKT pathway regulation, promoting the expression of collagen fibers.
Collapse
Affiliation(s)
- Liujun Zeng
- Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China
| | - Hui Peng
- Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Hunan University of Medicine General Hospital, China
| | - Leiye Sun
- Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China
| | - Huiqiao Yu
- Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China
| | - Yingfang Wu
- Centre of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Oral Cancer and Precancerous Lesions, Central South University, China.
| |
Collapse
|
36
|
Xia Q, Liu X, Zhong L, Qu J, Dong L. SMURF1 mediates damaged lysosomal homeostasis by ubiquitinating PPP3CB to promote the activation of TFEB. Autophagy 2025; 21:530-547. [PMID: 39324484 PMCID: PMC11849922 DOI: 10.1080/15548627.2024.2407709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024] Open
Abstract
The calcium-activated phosphatase PPP3/calcineurin dephosphorylates TFEB (transcription factor EB) to trigger its nuclear translocation and the activation of macroautophagic/autophagic targets. However, the detailed molecular mechanism regulating TFEB activation remains poorly understood. Here, we highlighted the importance of SMURF1 (SMAD specific E3 ubiquitin protein ligase 1) in the activation of TFEB for lysosomal homeostasis. SMURF1 deficiency prevents the calcium-triggered ubiquitination of the catalytic subunit of PPP3/calcineurin in a manner consistent with defective autophagic degradation of damaged lysosomes. Mechanically, PPP3CB/CNA2 plays a bridging role in the recruitment of SMURF1 by LGALS3 (galectin 3) upon lysosome damage. Importantly, PPP3CB increases the dissociation of the N-terminal tail (NT) and C-terminal carbohydrate-recognition domain (CRD) of LGALS3, which may promote the formation of open conformers in a PPP3CB dephosphorylation activity-dependent manner. In addition, PPP3CB is ubiquitinated at lysine 146 by the recruited SMURF1 in response to intracellular calcium stimulation. The K63-linked ubiquitination of PPP3CB enhances the recruitment of TFEB. Moreover, TFEB directly interacts with both PPP3CB and the regulatory subunit PPP3R1 which facilitate the conformational correction of TFEB for its activation for the transcription of TFEB-targeted genes. Altogether, our results highlighted a critical mechanism for the regulation of PPP3/calcineurin activity via its ubiquitin ligase SMURF1 in response to lysosomal membrane damage, which may account for a potential target for the treatment of stress-related diseases.Abbreviation AID: autoinhibitory domain; ATG: autophagy related; CD: catalytic domain; CRD: carbohydrate-recognition domain; CsA: cyclosporin A; DMSO: dimethyl sulfoxide; ESCRT: endosomal sorting complexes required for transport; GSK3B: glycogen synthase kinase 3 beta; LAMP1: lysosomal associated membrane protein 1; LGALS3: galectin 3; LLOMe: L-leucyl-L-leucine methyl ester; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; ML-SA1: mucolipin synthetic agonist 1; MTORC1: mechanistic target of rapamycin kinase complex 1; NT: N-terminal tail; PPP3CB: protein phosphatase 3 catalytic subunit beta; PPP3R1: protein phosphatase 3 regulatory subunit B, alpha; SMURF1: SMAD specific E3 ubiquitin protein ligase 1; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; VCP/p97: valosin containing protein; YWHA/14-3-3: tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein.
Collapse
Affiliation(s)
- Qin Xia
- Department of General Surgery, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xuan Liu
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lu Zhong
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- Department of General Surgery, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
- State Key Laboratory of Hearing and Balance Science and Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
37
|
Dev S, Asthana S, Singh P, Seth P, Banerjee C, Mukhopadhyay CK. Dopamine degrades ferritin by chaperone-mediated autophagy to elevate mitochondrial iron level in astroglial cells. Free Radic Biol Med 2025; 229:39-57. [PMID: 39818240 DOI: 10.1016/j.freeradbiomed.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Iron accumulation and mitochondrial dysfunction in astroglia are reported in Parkinson's disease (PD). Astroglia control iron availability in neurons in which dopamine (DA) synthesis is affected in PD. Despite their intimate relationship the role of DA in astroglial iron homeostasis is limited. Here we show that DA degrades iron storage protein ferritin in astroglial cells involving lysosomal proteolysis. Lysosomal ferritinophagy is mainly associated with macroautophagy; however, we revealed the involvement of chaperone-mediated autophagy (CMA) in DA-induced ferritin degradation. In CMA, cytosolic proteins containing a specific pentapeptide motif bind with HSC70 to be transported to lysosome mediated by LAMP2A. We identified the conserved pentapeptide motif in ferritin-H (Ft-H), mutations of which resulted loss of its interaction with HSC70. Pharmacological inhibitors of HSC70 or LAMP2/2A knockdown blocks DA-induced Ft-H degradation. DA also induces cytosolic cargo NCOA4 for ferritinophagy. We further reveal that DA promotes cathepsin B to lysis ferritin within the lysosome. Inhibitor of cathepsin B, knocking down of LAMP2, or HSC70 inhibitor attenuate DA-induced elevated mitochondrial iron level. Our results establish a direct role of DA on astroglial iron homeostasis and novel involvement of CMA in ferritin degradation in response to a biological stimulus. These results also may help in better understanding iron dyshomeostasis and mitochondrial dysfunction reported in PD.
Collapse
Affiliation(s)
- Som Dev
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India; Department of Biochemistry, All India Institute of Medical Sciences, Kalyani, West Bengal, India, 741245
| | - Somya Asthana
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pratibha Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pankaj Seth
- National Brain Research Centre, Manesar, Haryana, 122052, India
| | - Chayanika Banerjee
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Chinmay K Mukhopadhyay
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
38
|
Yang H, Gao S, Lu G, He J, Dong J, Zhang X, Liu L, Zhong K, Zha G, Han L, Guo S, Li H, Wang Y. SIRT5-mediated GLS and GDH desuccinylation attenuates the autophagy of bovine mammary epithelial cells induced by ammonia. Cell Signal 2025; 127:111570. [PMID: 39694127 DOI: 10.1016/j.cellsig.2024.111570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Sirtuin 5 (SIRT5) in mitochondria possesses a strong capacity for lysine desuccinylation, involving in various biological processes. Our previous research demonstrated that NH3 regulated autophagy dependent on SIRT5 in bovine mammary epithelial cells (bMECs). Interestingly, we discovered that SIRT5 reduced the content of NH3 and glutamate by inhibiting GLS activity in bMECs, the ratio of ADP/ATP also declined. In this study, we identified that SIRT5 interacted with endogenous GLS and GDH through Co-IP assay, but had no effect on endogenous GLS and GDH expression. SIRT5 made the succinylation levels of GLS and GDH significantly declined and resulted in the reduction of GLS and GDH activity. Next, the content of ammonia and glutamate, as well as the related autophagy markers were measured, we found that SIRT5 affected the glutamine metabolism, which attenuated ammonia release in MAC-T cells, accompanying with cellular autophagy decline, reducing the formation of autophagosome. Deletion of SIRT5 gene in MAC-T cells by means of CRISPR-cas9, we found the content of NH3 and glutamate increased, as well as autophagy promoted, which could be alleviated by SIRT5 overexpression. SIRT5 KO also resulted in increase of succinylation of GLS and GDH, as well as autophagy response in bMECs. Furthermore, SIRT5 promoted the maintenance of mitochondria homeostasis. Mechanistically, SIRT5 reduced ammonia release by modulating the succinylation levels and enzymatic activities of GLS and GDH in mitochondria and promoted the maintenance of mitochondria homeostasis, as well as further attenuated ammonia-stimulated autophagy in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Hanlin Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Shikai Gao
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Guangyang Lu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Junhui He
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Jinru Dong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Xinyi Zhang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Luya Liu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Guangming Zha
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Liqiang Han
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Shuang Guo
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Heping Li
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China.
| | - Yueying Wang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Key Laboratory of Veterinary Biotechnology of Henan Province, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China.
| |
Collapse
|
39
|
Stevens M, Wang Y, Bouley SJ, Mandigo TR, Sharma A, Sengupta S, Housden A, Perrimon N, Walker JA, Housden BE. Inhibition of autophagy as a novel treatment for neurofibromatosis type 1 tumors. Mol Oncol 2025; 19:825-851. [PMID: 39129390 PMCID: PMC11887668 DOI: 10.1002/1878-0261.13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/29/2024] [Accepted: 07/19/2024] [Indexed: 08/13/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder caused by mutation of the NF1 gene that is associated with various symptoms, including the formation of benign tumors, called neurofibromas, within nerves. Drug treatments are currently limited. The mitogen-activated protein kinase kinase (MEK) inhibitor selumetinib is used for a subset of plexiform neurofibromas (PNs) but is not always effective and can cause side effects. Therefore, there is a clear need to discover new drugs to target NF1-deficient tumor cells. Using a Drosophila cell model of NF1, we performed synthetic lethal screens to identify novel drug targets. We identified 54 gene candidates, which were validated with variable dose analysis as a secondary screen. Pathways associated with five candidates could be targeted using existing drugs. Among these, chloroquine (CQ) and bafilomycin A1, known to target the autophagy pathway, showed the greatest potential for selectively killing NF1-deficient Drosophila cells. When further investigating autophagy-related genes, we found that 14 out of 30 genes tested had a synthetic lethal interaction with NF1. These 14 genes are involved in multiple aspects of the autophagy pathway and can be targeted with additional drugs that mediate the autophagy pathway, although CQ was the most effective. The lethal effect of autophagy inhibitors was conserved in a panel of human NF1-deficient Schwann cell lines, highlighting their translational potential. The effect of CQ was also conserved in a Drosophila NF1 in vivo model and in a xenografted NF1-deficient tumor cell line grown in mice, with CQ treatment resulting in a more significant reduction in tumor growth than selumetinib treatment. Furthermore, combined treatment with CQ and selumetinib resulted in a further reduction in NF1-deficient cell viability. In conclusion, NF1-deficient cells are vulnerable to disruption of the autophagy pathway. This pathway represents a promising target for the treatment of NF1-associated tumors, and we identified CQ as a candidate drug for the treatment of NF1 tumors.
Collapse
Affiliation(s)
- Megan Stevens
- Living Systems InstituteUniversity of ExeterUK
- Department of Clinical and Biomedical ScienceUniversity of ExeterUK
| | - Yuanli Wang
- Living Systems InstituteUniversity of ExeterUK
- The First People's Hospital of QinzhouChina
| | | | - Torrey R. Mandigo
- Center for Genomic MedicineMassachusetts General HospitalBostonMAUSA
| | - Aditi Sharma
- Center for Genomic MedicineMassachusetts General HospitalBostonMAUSA
| | - Sonali Sengupta
- Living Systems InstituteUniversity of ExeterUK
- Department of Clinical and Biomedical ScienceUniversity of ExeterUK
| | - Amy Housden
- Living Systems InstituteUniversity of ExeterUK
| | - Norbert Perrimon
- Department of Genetics, Blavatnik InstituteHarvard Medical SchoolBostonMAUSA
- Howard Hughes Medical InstituteNew YorkNYUSA
| | - James A. Walker
- Center for Genomic MedicineMassachusetts General HospitalBostonMAUSA
- Cancer ProgramBroad Institute of MIT and HarvardCambridgeMAUSA
- Department of Neurology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Benjamin E. Housden
- Living Systems InstituteUniversity of ExeterUK
- Department of Clinical and Biomedical ScienceUniversity of ExeterUK
| |
Collapse
|
40
|
Jiang ZT, Chen J, Chen FY, Cheng YQ, Yao SY, Ma R, Li WB, Chen H, Guo DS. A FRET Autophagy Imaging Platform by Macrocyclic Amphiphile. Angew Chem Int Ed Engl 2025; 64:e202420793. [PMID: 39667942 DOI: 10.1002/anie.202420793] [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: 10/27/2024] [Revised: 11/24/2024] [Accepted: 12/12/2024] [Indexed: 12/14/2024]
Abstract
Autophagy is a ubiquitous process of organelle interaction in eukaryotic cells, in which various organelles or proteins are recycled and operated through the autophagy pathway to ensure nutrient and energy homeostasis. Although numerous fluorescent probes have been developed to image autophagy, these environment-responsive probes suffer from inherent deficiencies such as inaccuracy and limited versatility. Here, we present a modular macrocyclic amphiphile Förster Resonance Energy Transfer (FRET) platform (SC6A12C/NCM, SN), constructed through the amphiphilic assembly of sulfonatocalix[6]arene (SC6A12C) with N-cetylmorpholine (NCM) for lysosome targeting. The hydrophobic fluorophore BPEA (FRET donor) was entrapped within the inner hydrophobic phase and showed strong fluorescence emission. Attributed to the broad-spectrum encapsulation of SC6A12C, three commercially available organelle probes (Mito-Tracker Red, ER Tracker Red, and RhoNox-1) were selected as SC6A12C guests (FRET acceptors). During autophagy process, the formation of intracellular host-guest complexes leads to strong FRET signal, allowing us to visualize the fusion of mitochondria, endoplasmic reticulum, and Golgi apparatus with lysosomes, respectively. This study provides a versatile and accessible platform for imaging organelle autophagy.
Collapse
Affiliation(s)
- Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Jie Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Fang-Yuan Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Yuan-Qiu Cheng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Shun-Yu Yao
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Rong Ma
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Wen-Bo Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
| | - Hongzhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, 300071, Tianjin, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi, 844000, China
| |
Collapse
|
41
|
Guo N, Xia Y, He N, Zhang L, Liu J. IRGM Inhibits the AKT/mTOR Signaling Pathway by Interacting with TRIM21 to Alleviate Sepsis-Induced Acute Lung Injury. Inflammation 2025:10.1007/s10753-025-02265-w. [PMID: 39994091 DOI: 10.1007/s10753-025-02265-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025]
Abstract
Acute lung injury (ALI) is a severe complication of sepsis, and its underlying pathological mechanisms remain poorly understood. This study aims to investigate the role and mechanisms by which IRGM mediates autophagy through the regulation of the AKT/mTOR signaling pathway in sepsis-induced ALI. Initially, a sepsis-induced ALI mouse model was established using cecal ligation and puncture (CLP). Our results demonstrated that Irgm1 expression was significantly upregulated in the ALI model. Subsequently, Irgm1 was knocked down in vivo using AAV vectors, and changes in ALI symptoms were assessed. In vitro, a sepsis-induced ALI cell model was generated by stimulating A549 cells with lipopolysaccharide (LPS). The effects of IRGM overexpression on autophagy and apoptosis were evaluated, and its impact on the AKT/mTOR signaling pathway was analyzed. Furthermore, mass spectrometry and co-immunoprecipitation (COIP) experiments were conducted to explore the interaction between IRGM and TRIM21. In vivo results showed that Irgm1 knockout exacerbated CLP-induced ALI, as evidenced by a significant reduction in autophagic activity, increased apoptosis, and aberrant activation of the AKT/mTOR pathway. Further cellular experiments suggested that IRGM may enhance autophagy by inhibiting the AKT/mTOR signaling pathway, thereby attenuating LPS-induced cell damage. Additionally, COIP experiments revealed that IRGM interacts with TRIM21 to inhibit AKT/mTOR pathway activation, thereby promoting autophagy and mitigating sepsis-induced ALI. In conclusion, IRGM regulates autophagy through the AKT/mTOR signaling pathway and exerts protective effects in sepsis-induced ALI, suggesting that it may serve as a potential therapeutic target for sepsis-related ALI.
Collapse
Affiliation(s)
- Na Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yu Xia
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China
| | - Nannan He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China
| | - Lei Zhang
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou, Gansu Province, China.
| | - Jian Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu Province, China.
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou, Gansu Province, China.
| |
Collapse
|
42
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial Autophagic Dysregulation in Traumatic Brain Injury: Molecular Insights and Therapeutic Avenues. ACS Chem Neurosci 2025; 16:543-562. [PMID: 39920904 DOI: 10.1021/acschemneuro.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted condition that can result in cognitive and behavioral impairments. One aspect of TBI that has received increasing attention in recent years is the role of microglia, the brain-resident immune cells, in the pathophysiology of the injury. Specifically, increasing evidence suggests that dysfunction in microglial autophagy, the process by which cells degrade and recycle their own damaged components, may contribute to the development and progression of TBI-related impairments. Here, we unravel the pathways by which microglia autophagic dysregulation predisposes the brain to secondary damage and neurological deficits following TBI. An overview of the role of autophagic dysregulation in perpetuation and worsening of the inflammatory response, neuroinflammation, and neuronal cell death in TBI follows. Further, we have evaluated several signaling pathways and processes that contribute to autophagy dysfunction-mediated inflammation, neurodegeneration, and poor outcome in TBI. Additionally, a discussion on the small molecule therapeutics employed to modulate these pathways and mechanisms to treat TBI have been presented. However, additional research is required to fully understand the processes behind these underlying pathways and uncover potential therapeutic targets for restoring microglial autophagic failure in TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, California 92037, United States
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University, Sakaka 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research, New Delhi 201002, India
| | | |
Collapse
|
43
|
Gao K, Zhao Y, Si M, Zhang B, Wang Z, Chen H, Lin P, Wang A, Jin Y. ERS regulates endometrial epithelial cell autophagy through XBP1s-mediated activation of the PI3K/AKT pathway. Sci Rep 2025; 15:5943. [PMID: 39966508 PMCID: PMC11836410 DOI: 10.1038/s41598-024-84461-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/23/2024] [Indexed: 02/20/2025] Open
Abstract
Autophagy is a fundamental cellular activity involved in the renewal of cellular components, occurring primarily in cells subjected to physiological remodeling or pathological stimuli. The occurrence of autophagy is closely related to the endoplasmic reticulum (ER), and ER stress (ERS) occurs when ER homeostasis is disrupted. The current study aimed to analyze the molecular mechanisms underlying the effects of ERS on autophagy in goat endometrial epithelial cells (gEECs). We found that rapamycin (an autophagy inducer) induced autophagy and ERS in a time-dependent manner in gEECs which was accompanied by significantly increased expression of the autophagy-related genes ATG5, the LC3II/LC3I and ERS-related genes GRP78, IRE1, ATF6, and XBP1s. PI3K and AKT protein phosphorylation was significantly reduced during gEECs autophagy. Interestingly, TG (ERS activator) significantly inhibited the expression of ATG5 and the LC3II/LC3I and significantly promoted expression of SQSTM1, whereas the ERS inhibitor 4-PBA showed the opposite results. Surprisingly, XBP1s knockdown inhibited the effects of TG. Furthermore, XBP1s overexpression significantly inhibited autophagy whereas XBP1s knockdown increased ATG5 expression and the LC3II/LC3I and decreased SQSTM1 expression in gEECs. Specifically, XBP1s overexpression significantly promoted PI3K and AKT protein phosphorylation while treatment with LY294002 (PI3K/AKT pathway inhibitor) significantly reversed the effect. Similarly, PI3K/AKT pathway activation was significantly inhibited following XBP1s knockdown whereas treatment with SC79 (PI3K/AKT pathway activator) showed the opposite results. Taken together, our data suggest that interactions between ERS and autophagy exist in gEECs. XBP1s, the key effector of ERS, inhibits autophagy in gEECs by promoting the PI3K/AKT pathway in gEECs. These results may contribute to the treatment and prevention of uterine diseases.
Collapse
Affiliation(s)
- Kangkang Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yiteng Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Mengqi Si
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Beibei Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zongjie Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Pengfei Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
44
|
Ren Z, Liu X, Lv G, Wang X, Wang J, Wu W, Li X, Wang J. Mitochondrial Localization of Antioxidant Nanodrug Suppresses Ocular Inflammation by Alleviating Oxidative Stress on Cells. Chemistry 2025; 31:e202402441. [PMID: 39833114 DOI: 10.1002/chem.202402441] [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: 06/26/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 01/22/2025]
Abstract
External environments (e. g., pollutants, irritants, ultraviolet radiation, etc) probably activate oxidative stress on the ocular surface, further leading to inflammatory responses and cellular apoptosis. For treating ophthalmic diseases, one of the strategies is to regulate oxidative stress through antioxidants. Here we conjugate a polyphenolic antioxidant drug (i. e., caffeic acid) with a small peptide of protein tag to generate a peptide-drug conjugate as a nanodrug. With a self-assembled ability to form nanoparticles, the nanodrug mainly enters human corneal epithelial cells (HCEC) by caveolin-mediated endocytosis, succeeds lysosomal escape, and achieves mitochondrial localization of caffeic acid. Revealed by free radical scavenging experiments, the nanodrug shows considerable antioxidant capacities. In mouse leukemia cells of monocyte macrophage (RAW 264.7) induced by lipopolysaccharide (LPS), the nanodrug inhibits various pro-inflammatory cytokines (e. g., NO, IL-6, and TNF-α) by up-regulating the expression of anti-apoptotic protein (e. g., Bcl-2). As an investigation of alleviating oxidative stress by the mitochondrial localization of antioxidant, this work may establish an experimental foundation for the regulation of cellular redox balance, as well as provide different insights for the clinical development of antioxidant drug delivery systems in the treatment of ocular disease.
Collapse
Affiliation(s)
- Zhibin Ren
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Xiaoying Liu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Guanghao Lv
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Xiaiting Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Jingli Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Wei Wu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Xingyi Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| | - Jiaqing Wang
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology and Optometry & Biomedical Engineering, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China
| |
Collapse
|
45
|
Bhadauriya P, Onkar A, Nagarajan K, Angamuthu Karuppusamy K, Ganesh S, Agarwal S. Glycogen synthase is required for heat shock-mediated autophagy induction in neuronal cells. Biol Open 2025; 14:BIO061605. [PMID: 39912200 PMCID: PMC11876841 DOI: 10.1242/bio.061605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Autophagy is an essential cellular process that facilitates the degradation of aggregated proteins and damaged organelles to maintain cellular homeostasis and promote cell survival. Recent studies have indicated a direct role for glycogen synthase (GS) in activating neuronal autophagy and in conferring protection against cytotoxic misfolded proteins. Since heat shock induces protein misfolding and autophagy is an essential component of the heat shock response that clears the misfolded proteins, we looked at the possible role of GS in heat shock response pathways in neuronal cells. We demonstrate an increase in the activity and level of GS and a concomitant increase in the glycogen level during the heat shock and post-heat shock recovery period. These changes had a direct correlation with autophagy induction. We further demonstrate that heat shock transcription factor 1 regulates the level and activation of GS during heat shock and that GS is essential for the induction of autophagy during heat stress in neuronal cells. Intriguingly, the partial knock-down of GS led to increased death due to heat shock in neuronal cells and Drosophila. Our study offers a novel insight into the role of GS and glycogen metabolic pathways in heat shock response in neuronal cells.
Collapse
Affiliation(s)
- Pratibha Bhadauriya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Akanksha Onkar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Kamali Nagarajan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | | | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur 208016, India
- Gangwal School of Medical Sciences and Technology, Indian Institute of Technology, Kanpur 208016, India
| | - Saloni Agarwal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| |
Collapse
|
46
|
Tie J, Guo J, Huang Y, Huang Z, Yan Z, Yuan J, Shen X, Wang J. A novel lncRNA enhances autophagy to suppress extracellular matrix via modulating TP53INP1 in human trabecular meshwork cells under oxidative stress. Sci Rep 2025; 15:5049. [PMID: 39934637 DOI: 10.1038/s41598-024-81300-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Primary open-angle glaucoma (POAG) can result in irreversible blindness. As an important etiological factor, oxidative stress can elicit extraordinary increase of extracellular matrix (ECM) in trabecular meshwork-schlemm canal, to increase aqueous humor outflow resistance and elevate intraocular pressure. Although autophagy plays an important role in clearing ECM, the functions of long non-coding RNAs (lncRNAs) in autophagy induced by oxidative stress in human trabecular meshwork cells (HTMCs) remain unclear. In our study, oxidative stress induced the expression of ECM and autophagy in TMCs after H2O2 treatment. Meanwhile, a novel lncRNA ENST00000523905 and tumor protein 53-induced nuclear protein 1 (TP53INP1) were elevated in TMCs treated with H2O2. Similar to treatment with 3-MA (an inhibitor of autophagy), knocking-down the expression of TP53INP1 or ENST00000523905 could suppress the autophagy of TMCs induced by H2O2, which increased the level of ECM. Furthermore, the inhibition of ENST00000523905 decreased the expression of TP53INP1. ENST00000523905 could recruit and directly bind with CCAAT/enhancer (C/EBPβ), which can promote the expression of TP53INP1. Taken together, our findings demonstrated that ENST00000523905 may increase autophagy via enhancing TP53INP1 expression through binding with C/EBPβ, resulting in oxidative stress-induced decrease in ECM in HTMCs.
Collapse
Affiliation(s)
- Jinjun Tie
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
- Department of Ophthalmology, The Affiliated Hospital of Gui Zhou Medical University, Guiyang, Guizhou, China
| | - Junhong Guo
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Yijia Huang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Zihan Huang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Zhichao Yan
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Jiemei Yuan
- Department of Ophthalmology, The Affiliated Hospital of Gui Zhou Medical University, Guiyang, Guizhou, China
| | - Xiaoli Shen
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China.
| | - Jiantao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China.
| |
Collapse
|
47
|
Huang H, Liang X, Li S, Yan Y, Li S, Qiu C, Ye Z, Zhu Y, Shen D, Lin Y, Wang L, Chen N, Yao Y, Zhao X, Wu F, Shi X, Kou L, Chen R, Yao Q. Chondrocyte-targeted bilirubin/rapamycin carrier-free nanoparticles alleviate oxidative stress and modulate autophagy for osteoarthritis therapy. J Control Release 2025; 378:517-533. [PMID: 39701459 DOI: 10.1016/j.jconrel.2024.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic disease, characterized by the destruction of joint cartilage and synovitis, affects over 7 % of people worldwide. Disease-modifying treatments for OA still face significant challenges. Chondrocytes, as the exclusive cellular component of articular cartilage, play a pivotal role in synthesizing the intricate matrix of cartilage, thereby assuming a critical responsibility in facilitating its renewal and repair processes. However, oxidative stress within chondrocytes and subsequent apoptotic cell death plays significant roles in the progression of OA. Therefore, targeting apoptosis inhibition and mitigation of oxidative stress in chondrocytes represents a promising therapeutic strategy for OA. This study develops a type II collagen-targeting peptide (WYRGRLC) modified bilirubin/rapamycin carrier-free nanoparticle (PP/BRRP) and evaluate its therapeutic potential for OA. The PP/BRRP system exhibits remarkable chondrocyte-targeting ability, enabling the rupture of highly oxidized chondrocytes and subsequent release of bilirubin and rapamycin. This dual payload effectively scavenges reactive oxygen species, triggers autophagy, and suppresses the mTOR pathway, thereby augmenting anti-inflammatory and anti-apoptotic effects. The in vivo experiments further validate the retention and therapeutic efficacy of PP/BRRP in rat joints affected by OA. Overall, PP/BRRP exhibits significant potential for intervention and treatment of OA.
Collapse
Affiliation(s)
- Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xindan Liang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shengjie Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yuqi Yan
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shize Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhanzheng Ye
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yixuan Zhu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Dingchao Shen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinhao Lin
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Luhui Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Nuo Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yinsha Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Fugen Wu
- Department of Pediatric, The First People's Hospital of Wenling, Taizhou, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Pediatrics Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
48
|
Liu D, Wang H, Li J, Sheng S, Wang S, Tian Y. Non-lethal sonodynamic therapy mitigates hypertensive renal fibrosis through the PI3K/AKT/mTORC1-autophagy pathway. Sci Rep 2025; 15:4534. [PMID: 39915557 PMCID: PMC11802789 DOI: 10.1038/s41598-025-86973-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
Hypertension constitutes a significant public health concern, characterized by a high incidence and mortality rate. Hypertensive kidney disease is a prevalent complication associated with hypertension and is the second leading cause of end-stage renal disease (ESRD). Renal fibrosis linked to hypertension has emerged as the third leading cause of disease in dialysis patients. Autophagy activity is crucial for maintaining homeostasis, vitality, and physiological function of kidney cells, while also protecting the kidneys from fibrosis. The deficiency of autophagy will increase the sensitivity of the kidney to the damage, leading to impaired renal function, accumulation of damaged mitochondria and more severe of renal fibrosis. However, enhancing autophagy by activating the PI3K/AKT, AMPK, and mTOR pathways, improves podocyte injury and renal pathological changes, and ameliorates renal function. Current clinical interventions aimed at halting or reversing renal fibrosis in hypertensive patients are notably limited in their efficacy. Here, we present Non-lethal Sonodynamic Therapy (NL-SDT), in which ultrasound is used to activate locally sonosensitizers, thereby stimulating the production of reactive oxygen species for the purpose of modulating cell function or fate, as a novel methodology to inhibit progression of hypertensive renal fibrosis.To confirm whether NL-SDT can reduce hypertensive renal fibrosis and its mechanism. The mice model of hypertensive renal fibrosis was established by using osmotic minipumps (Alzet model 2004, Cupertino, CA) equipped with angiotensin-II (Ang II). The pumps were implanted in mice, ensuring constant infusion of Ang II at a dose of 1.0 µg/kg per minute for 4 weeks. The mice were exposed to 0.4 W/cm2 intensity ultrasonic radiation for 15 min at 4 h post injection of sinoporphyrin sodium (DVDMS) (4 mg/kg) into the caudal vein was repeated weekly for 4 treatments. The kidney from mice was stained with masson's trichrome staining for collagen fiber expression, while alpha-smooth muscle actin (α-SMA) expression was determined via immunohistochemical staining. The protein levels of fibrosis parameters (α-SMA, collagen I, vimentin), pathway-related proteins (PI3K, AKT, mTORC1) and autophagy-related protein LC3B were determined using western blotting. Intracellular reactive oxygen species (ROS) levels were detected using DCFH-DA probe. Immunofluorescence was also used to observe the expression of α-SMA and E-cadherin in cells. Pathway-related protein inhibitors (the autophagy-related inhibitor 3-methyladenine (3-MA), chloroquine (CQ), ROS inhibitor N-acetyl-L-cysteine (NAC) were applied, and autophagosome changes were observed under transmission electron microscopy. Immunofluorescence was used to observe LC3 spot formation within cells.We obtained the following results via animal and cellular research. In vivo, (1) The collagen area of renal tissue was increased significantly in Ang II group (50.6%). The positive expression of α-SMA was increased significantly (37.8%). (2) The collagen area decreased after NL-SDT treatment (34.8%). The expression of α-SMA was decreased too (48.9%). The expression of LC3B increased in NL-SDT group. (3) The effect of NL-SDT on reducing renal fibrosis can be changed by rapamycin and CQ. In vitro. (1) The expression of α-SMA, collagen I and vimentin were increased significantly in TGF-β1-induced NRK-52E cells. (2) The increase of autophagosomes was observed in TGF-β1-induced NRK-52E cells after NL-SDT. The levels of ROS were increased after NL-SDT (24.8%). The effect of NL-SDT on autophagy was reversed after administration of NAC. The expression of PI3K, P-AKT and P-mTORC1 was decreased in TGF-β1-induced NRK-52E cells after NL-SDT. NL-SDT inhibited the transition of epithelial cells into myofibroblasts by activating PI3K-AKT-mTORC1-autophagy pathway in TGF-β1-induced NRK-52E cells. (3) The administration of the pathway inhibitors showed a reciprocal effect on NL-SDT-inhibited epithelial-mesenchymal transition (EMT).(1) NL-SDT reduced blood pressure temporarily in mice model of hypertensive renal fibrosis induced by Ang II. (2) NL-SDT alleviated renal fibrosis in mice model of hypertensive renal fibrosis induced by Ang II. (3) NL-SDT promoted autophagy by inhibiting PI3K-AKT-mTORC1 signaling pathway and alleviated renal fibrosis in mice model of hypertensive renal fibrosis induced by Ang II. NL-SDT is a non-invasive and efficacious regimen to inhibit renal fibrosis. It may be a new approach for clinical treatment of renal fibrosis, delaying or reducing the occurrence of ESRD.
Collapse
Affiliation(s)
- DanDan Liu
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China
| | - Hui Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China
| | - Jialong Li
- Department of Pathophysiology and Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, 150086, PR China
| | - Siqi Sheng
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China
| | - Shu Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China.
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, 150001, PR China.
| |
Collapse
|
49
|
Li J, Zhao Y, Wu X, Zou Y, Liu Y, Ma H. Choline kinase alpha regulates autophagy-associated exosome release to promote glioma cell progression. Biochem Biophys Res Commun 2025; 746:151269. [PMID: 39778250 DOI: 10.1016/j.bbrc.2024.151269] [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: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Glioma is the most common primary intracranial malignant tumor in adults, with a poor prognosis. Exosomes released by tumor cells play a crucial role in tumor development, metastasis, angiogenesis, and other biological processes. Despite this significance, the precise molecular mechanisms governing exosome secretion and their impact on tumor progression remain incompletely understood. While Choline Kinase Alpha (CHKA) has been implicated in promoting various types of tumors, its specific role in glioma pathogenesis remains unclear. Our study initially demonstrates that CHKA enhances the proliferation, migration, and invasion abilities of glioma cells. Interestingly, CHKA also stimulates the release of exosomes from glioma cells. Mechanistically, reduced CHKA expression hampers exosome secretion by elevating autophagy levels in gliomas, whereas counteracting the autophagy elevation resulting from CHKA downregulation restores the release of exosomes. Notably, exosomes derived from glioma cells with normal CHKA expression exhibit a greater capacity to promote glioma progression compared to those derived from cells with low CHKA expression. Overall, our findings suggest that CHKA modulates exosome secretion via an autophagy-dependent pathway, thereby facilitating the proliferation, migration, and invasion of glioma cells.
Collapse
Affiliation(s)
- Jialin Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yang Zhao
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiao Wu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yourui Zou
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yang Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hui Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
50
|
Rost-Roszkowska M, Urbisz A, Małota K, Wilczek G, Serda M, Skonieczna M. Investigation of potential cytotoxicity of a water-soluble, red-fluorescent [70]fullerene nanomaterial in Drosophila melanogaster. Nanotoxicology 2025; 19:1-16. [PMID: 39736806 DOI: 10.1080/17435390.2024.2445250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025]
Abstract
Fullerenes (C60, C70) as carbon nanomaterials can enter the environment through natural processes and anthropogenic activities, while synthetic fullerenes are commonly used in medicine in targeted therapies in association with antibodies, or anticancer and antimicrobial drugs. As the nanoparticles, they can pass through cell membranes and organelles and accumulate in the entire cytoplasm. The red-fluorescent, water-soluble [70]fullerene derivative C70-OMe-ser, which produces reactive oxygen species upon illumination with an appropriate wavelength, passed into the cytoplasm of the middle region in the Drosophila melanogaster digestive system. To determine whether [70]fullerene nanomaterials that produce fluorescence after entering the cell cytoplasm will hurt its homeostasis, it is necessary to investigate the activation of degenerative and possibly regenerative processes. In vivo, studies on the model species D. melanogaster may help to elucidate whether the water-soluble [70]fullerene derivative that produces fluorescence can still be considered among the most promising nanomaterials. The experiment involved feeding insects ad libitum with yeast paste supplemented with 40 µg of fullerenes/mL for 1 week and 1 month. Thus, adult females and males of D. melanogaster were divided into control (CWM, CWF, CMM, and CMF) and experimental groups (FWM, FWF, FMM, and FMF). The quantitative and qualitative analysis enabled the presentation of the effects of the water-soluble [70]fullerene derivatives on cell proliferation and degeneration. Our study presented that [70]fullerene derivative showed a cytoprotective effect and activated cell proliferation. Therefore, we could conclude that analyzed carbon nanomaterials seemed to be safe for the cells into which they have penetrated.
Collapse
Affiliation(s)
- Magdalena Rost-Roszkowska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Anna Urbisz
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Karol Małota
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Grażyna Wilczek
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Maciej Serda
- Institute of Chemistry, University of Silesia in Katowice, Katowice, Poland
| | - Magdalena Skonieczna
- Department of Systems Engineering and Biology, Silesian University of Technology, Faculty of Automatic Control, Electronics and Computer Science, Gliwice, Poland
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|