1
|
Ginet V, Depierre P, Puyal J. Neuronal autosis: the self-destructive side of autophagy involved in hypoxic-ischemic neuronal death. Neural Regen Res 2025; 20:3517-3518. [PMID: 39589723 PMCID: PMC11974643 DOI: 10.4103/nrr.nrr-d-24-00831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/13/2024] [Accepted: 10/11/2024] [Indexed: 11/27/2024] Open
Affiliation(s)
- Vanessa Ginet
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
- Clinic of Neonatology, Department of Women, Mother and Child, University Hospital Center of Vaud, Lausanne, Switzerland
| | - Pauline Depierre
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
- CURML, University Center of Legal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
2
|
García-Juan M, Villa M, Benito-Cuesta I, Ordóñez-Gutiérrez L, Wandosell F. Reassessing the AMPK-MTORC1 balance in autophagy in the central nervous system. Neural Regen Res 2025; 20:3209-3210. [PMID: 39715086 PMCID: PMC11881726 DOI: 10.4103/nrr.nrr-d-24-00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Marta García-Juan
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mario Villa
- Fisiología Animal, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Irene Benito-Cuesta
- Department of Clinical Neuroscience, CMM Karolinska Universitetssjukhuset Solna, Stockholm, Sweden
| | - Lara Ordóñez-Gutiérrez
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Universidad Complutense de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Nicolas Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
3
|
Sharma C, Kim S, Eo H, Kim SR. Recovery of the injured neural system through gene delivery to surviving neurons in Parkinson's disease. Neural Regen Res 2025; 20:2855-2861. [PMID: 39610091 PMCID: PMC11826474 DOI: 10.4103/nrr.nrr-d-24-00724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/22/2024] [Accepted: 09/11/2024] [Indexed: 11/30/2024] Open
Abstract
A critical unaddressed problem in Parkinson's disease is the lack of therapy that slows or hampers neurodegeneration. While medications effectively manage symptoms, they offer no long-term benefit because they fail to address the underlying neuronal loss. This highlights that the elusive goals of halting progression and restoring damaged neurons limit the long-term impact of current approaches. Recent clinical trials using gene therapy have demonstrated the safety of various vector delivery systems, dosages, and transgenes expressed in the central nervous system, signifying tangible and substantial progress in applying gene therapy as a promising Parkinson's disease treatment. Intriguingly, at diagnosis, many dopamine neurons remain in the substantia nigra, offering a potential window for recovery and survival. We propose that modulating these surviving dopamine neurons and axons in the substantia nigra and striatum using gene therapy offers a potentially more impactful therapeutic approach for future research. Moreover, innovative gene therapies that focus on preserving the remaining elements may have significant potential for enhancing long-term outcomes and the quality of life for patients with Parkinson's disease. In this review, we provide a perspective on how gene therapy can protect vulnerable elements in the substantia nigra and striatum, offering a novel approach to addressing Parkinson's disease at its core.
Collapse
Affiliation(s)
- Chanchal Sharma
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
| | - Sehwan Kim
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
| | - Hyemi Eo
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
| | - Sang Ryong Kim
- School of Life Science, Kyungpook National University, Daegu, Korea
- BK21 FOUR KNU Creative BioResearch Group Kyungpook National University, Daegu, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, Korea
| |
Collapse
|
4
|
Liu Z, Chu A, Bai Z, Yang C. Nobiletin ameliorates monosodium urate-induced gouty arthritis in mice by enhancing AMPK/mTOR-mediated autophagy to inhibit NF-κB/NLRP3 inflammasome activation. Immunol Lett 2025; 274:106982. [PMID: 39965668 DOI: 10.1016/j.imlet.2025.106982] [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: 07/23/2024] [Revised: 01/06/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Gouty arthritis (GA) is a common rheumatic disease caused by the release of monosodium urate crystal (MSU) deposits into joint space. Nobiletin is a polymethoxylated flavonoid isolated from citrus fruits and has many beneficial activities. This study aimed to elucidate the therapeutic efficacy of nobiletin in GA and to reveal its potential mechanisms. METHODS Phorbol-12-myristate-13-acetate (PMA)-differentiated THP-1 macrophages were primed with lipopolysaccharide (LPS) and then stimulated with MSU crystals in the presence or absence of nobiletin. Cell viability as well as the levels of proinflammatory cytokines, pathway-related proteins, NLRP3 inflammasomes, and autophagy-related proteins were evaluated. MSU was used to induce GA in mice. Hematoxylin-eosin staining was conducted to assess histological morphology changes. Immunofluorescence staining was performed to measure LC3 expression in THP-1 cells and ankle joint tissues. RESULTS For in vitro analysis, nobiletin reduced LPS and MSU-induced cell viability inhibition. Additionally, nobiletin inhibited inflammation and NF-κB/NLRP3 pathway in THP-1 cells. Moreover, nobiletin inhibited the activation of NLRP3 inflammasome by promoting AMPK/mTOR-mediated autophagy. For in vivo analysis, nobiletin attenuated MSU-induced GA in mice. Additionally, nobiletin suppressed inflammation and NF-κB/NLRP3 pathway and promoted tissue autophagy in GA mice. CONCLUSION Nobiletin prevents MSU-induced GA in mice by inhibiting NF-κB/NLRP3 inflammasome activation through AMPK/mTOR-mediated autophagy.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Rheumatology and Immunology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuchang District, Wuhan, Hubei 430060, China
| | - Aichun Chu
- Department of Rheumatology and Immunology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuchang District, Wuhan, Hubei 430060, China
| | - Zhiqian Bai
- Department of Rheumatology and Immunology, Wuhan University, Renmin Hospital, 238 Jiefang Road, Wuchang District, Wuhan, Hubei 430060, China
| | - Chao Yang
- Department of Orthopedics, Maternal and Child Health Hospital of Hubei Province, No 745 Wuluo Road, Hongshan District, Wuhan, Hubei 430070, China.
| |
Collapse
|
5
|
Franconi F, Lodde V, Capobianco G, Criscione M, Montella A, Campesi I. Effects of maternal smoking on inflammation, autophagy/mitophagy, and miRNAs in endothelial cells: Influence of newborn sex. Eur J Pharmacol 2025; 998:177648. [PMID: 40252896 DOI: 10.1016/j.ejphar.2025.177648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Maternal smoking (MS) during pregnancy is linked to well-documented adverse health effects for the mother and foetus, however the role of fetal sex was largely overlooked. Primary cultures of male and female human umbilical vein endothelial cells (MHUVECs and FHUVECs, respectively) were used. IL-6, IL-8, and TNF-α levels were measured in HUVECs supernatant. The expression of genes and proteins (oestrogen receptors (ERs), Hsp90, Beclin-1, p62, LC3, LAMP-1 and Parkin), as well as the expression of miR-29a-3p, miR-29b-3p, miR-126-3p, miR-133a-3p, and miR-146a-5p were analysed in cells obtained from foetuses born to non-smoking and smoking mothers. In HUVECs from foetuses born to non-smoking mothers, Beclin-1 protein was higher in MHUVECs (1.8 fold increase), while Parkin, Hsp90 proteins, and miR-146a-5p were elevated in FHUVECs (2.2, 2.6, and 2.1 fold increase, respectively), with no other significant differences. MS amplified these sex differences, with specific effects based on foetus sex. FHUVECs obtained from foetus born to smoking mothers showed higher levels of IL-8 (1399.36 ± 123.96 pg/ml for FHUVECs vs 655.11 ± 215.94; pg/ml for MHUVECs; P < 0.001), Hsp90 gene and protein (3.3 and 2.6 fold increase), and ERβ protein and Beclin-1 gene (2.1, and 4.9 fold increase), and lower levels of miR-29b-3p, miR-133a-3p, and miR-146a-5p than MHUVECs (0.27, 0.68, and 0.1 fold change, respectively). This study shows that primary HUVECs from fetuses born to smoking mothers retain a memory of smoking effects, with sex differences in gene expression, miRNA profiles, and autophagic responses, suggesting that maternal smoking impacts endothelial cell physiology in a sex-dependent manner.
Collapse
Affiliation(s)
- Flavia Franconi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100, Sassari, Italy
| | - Valeria Lodde
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy; Gynecologic and Obstetric Clinic, AOU, Viale San Pietro 12, 07100, Sassari, Italy
| | - Massimo Criscione
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100, Sassari, Italy; Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy.
| |
Collapse
|
6
|
He Z, Xu Y, Zhang Y, Jin M, Sun Y, Tang F, Qiu C, Junior AMA, Cai Y, Xu X, Chen X, Chen K, Xiang G, Xiao J, Wang J, Wang J, Chen B. Betulinic acid enhances autopahgy to promote microglial M2 polarization and alleviate inflammation via AMPK-HDAC5-KLF2 signaling pathways in spinal cord injury. Int Immunopharmacol 2025; 158:114889. [PMID: 40388862 DOI: 10.1016/j.intimp.2025.114889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Spinal cord injury (SCI) leads to neuroinflammation and activates microglia, which are crucial contributors to neurological deficits. Betulinic acid (BA), a naturally occurring pentacyclic triterpenoid, has demonstrated effectiveness in treating inflammatory and neurological disorders. This study aims to explore the potential role and underlying mechanism of BA in modulating microglial activation and inflammation in the context of SCI. Using a mouse SCI model, we assessed motor recovery via Basso Mouse Scale (BMS) and neuronal survival via H&E/Nissl staining. Western blotting, qPCR, immunofluorescence, and flow cytometry were employed to analyze microglial polarization, autophagy, and AMPK-HDAC5-KLF2 signaling in vivo and in LPS-stimulated BV2 cells. Our findings reveal that BA significantly enhances functional recovery and reduces neuronal apoptosis following SCI. Furthermore, BA facilitates the phenotypic transition of microglia from the M1 to M2 phenotype, thereby decreasing inflammatory factors in both the SCI model and LPS-stimulated BV2 cells. BA treatment restores the disrupted autophagy flux in microglia induced by SCI or LPS, which in turn mitigates M1 polarization and inflammation. Mechanistically, BA restores autophagy flux by activating the AMPK-HDAC5-KLF2 axis, thereby shifting microglia from pro-inflammatory M1 to anti-inflammatory M2 phenotype.
Collapse
Affiliation(s)
- Zili He
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yitie Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yu Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Mengqi Jin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Yinuo Sun
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Fangying Tang
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chuangqi Qiu
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Abass Mashud Akinfemi Junior
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yunhao Cai
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaodan Xu
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xianghang Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China
| | - Kongbin Chen
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Guangheng Xiang
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Jian Xiao
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China; Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Jian Wang
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Jing Wang
- Department of Wound Healing, Department of Spine Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Baoyi Chen
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
7
|
Xing QC, Chen J, Liu Z, Li WC, Liu X, Li W. Autophagy in Schwann cells: A potential pharmacotherapeutic target in diabetic peripheral neuropathy. World J Diabetes 2025; 16:105709. [DOI: 10.4239/wjd.v16.i6.105709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/27/2025] [Accepted: 05/16/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes and is characterized by sensory and motor impairments resulting from neural injury. Schwann cells (SCs), which are important for peripheral nerve function, are compromised under hyperglycemic conditions, leading to impaired axonal regeneration and demyelination. Autophagy, a cellular degradation process, is essential for SC function and significantly influences DPN progression. This article highlights the significance of autophagy in SCs and its potential as a pharmacotherapeutic target in DPN. We discuss the mechanisms of autophagy in SCs, including the mammalian target of rapamycin, adenosine monophosphate-activated protein kinase, and phosphatase and tensin homolog-induced putative kinase/parkin pathways, and their dysregulation in DPN. This article also examines various natural products and chemical agents that modulate autophagy and enhance the efficacy of DPN treatment. These agents target key signaling pathways, such as adenosine monophosphate-activated protein kinase/mammalian target of rapamycin and demonstrate potential in promoting nerve regeneration and restoring SC function. The roles of exosomes, long non-coding RNA, and proteins in the regulation of autophagy have also been explored. In conclusion, targeting autophagy in SCs is a promising strategy for DPN treatment and offers new insights into therapeutic interventions. Further research is warranted to fully exploit these targets for clinical applications.
Collapse
Affiliation(s)
- Qi-Chang Xing
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Jia Chen
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Zheng Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Wen-Can Li
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| | - Wei Li
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan 411100, Hunan Province, China
| |
Collapse
|
8
|
Domingues C, Jarak I, Matos A, Veiga F, Vitorino C, Dourado M, Figueiras A. Unraveling rosmarinic acid anticancer mechanisms in oral cancer malignant transformation. Eur J Pharmacol 2025; 997:177466. [PMID: 40064225 DOI: 10.1016/j.ejphar.2025.177466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Oral squamous cell carcinoma (OSCC) is expected to rise ca. 40 % by 2040. Rosmarinic acid (RA) has been recognized for its anticancer properties, although its role in OSCC has been neglected. This work exploits the activity of RA in 2D and 3D models of OSCC cells to compel a roadmap for its anticancer properties. The results demonstrated that RA significantly reduced cell mass and metabolic activity in a dose, time, and cell-type-dependent manner, predominantly in highly-invasive OSCC, without compromising normal mucosa in therapeutic doses. RA decreased mitochondria membrane potential and increased redox state, which was corroborated by pioneering observations on the metabolome landscape of OSCC cells (glutathione reduction and acetate and fumarate release). RA triggered autophagy, upregulating BNIP3 and BCNL1 and downregulating BIRC5. The upregulation of CADM1 and downregulation of VIM, CADM2, SNAIL1, and SOX9 highlighted the modulation of epithelial-mesenchymal transition and the remodeling of the extracellular matrix by the downregulation of MMP-2 and MMP-9. RA interacts with P-glycoprotein with the highest docking score of -6.4 kcal/mol. The HSC-3 cell surface charge decreased after RA treatment (-22.6 ± 0.3 mV vs. -26.3 ± 0.3 mV, p < 0.0001), suggesting a reversion of cell polarity and the impairment of invasion. RA also shrank the growth and the metabolic activity of multicellular tumor spheroids. Its modest protein binding with human saliva sheds light on its administration by the oromucosal route. Overall, this work supports the need for further research on the anticancer potential of RA in OSCC, either in monotherapy, combined with conventional treatments, or conveyed in nanosystems.
Collapse
Affiliation(s)
- Cátia Domingues
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; REQUIMTE/LAQV, Drug Development and Technologies Laboratory, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ivana Jarak
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Ana Matos
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; Chemical Engineering and Renewable Resources for Sustainability, CERES, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Francisco Veiga
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; REQUIMTE/LAQV, Drug Development and Technologies Laboratory, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Carla Vitorino
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Marília Dourado
- Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal; REQUIMTE/LAQV, Drug Development and Technologies Laboratory, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
9
|
Casetti R, Ciccosanti F, Lamsira HK, Pinnetti C, Mazzotta V, Ciolfi S, Sacchi A, Amendola A, Ippolito G, Piacentini M, Nardacci R. Autophagy is influenced by vitamin D 3 level in people with HIV-1. Biol Direct 2025; 20:69. [PMID: 40514685 DOI: 10.1186/s13062-025-00660-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Autophagy is the primary catabolic process responsible for degrading intracellular components and potentially harmful cytosolic entities by delivering them to lysosomes. Notably, this mechanism is crucial for controlling intracellular pathogens, with significant implications for both innate and adaptive immunity. In the context of HIV-1 infection, emerging evidence suggests that autophagy contributes to immune responses against the virus. Various compounds can modulate autophagy, among which vitamin D₃ is particularly effective due to its ability to prevent inflammation and slow HIV-1 disease progression. Indeed, vitamin D₃ contributes to regulating both innate and adaptive immunity, thereby modulating antiviral and antibacterial inflammatory responses. Importantly, vitamin D₃ deficiency is highly prevalent among people with HIV (PWH) and has been associated with an increased risk of severe disease progression. RESULTS In this study, we investigated the relationship between serum vitamin D₃ levels and the expression of autophagy markers in peripheral blood mononuclear cells from different categories of PWH: PWH under antiretroviral therapy (ART) with either normal vitamin D₃ levels or hypovitaminosis, and treatment-naïve PWH with either normal vitamin D₃ levels or hypovitaminosis. Our results show that low vitamin D₃ blood levels is associated with lower expression of the main factors involved in the autophagy mechanism, particularly in treatment-naïve PWH. CONCLUSIONS Our findings suggest that normal blood level of vitamin D₃ may play a crucial role in promoting autophagy in PWH. The observed differences in autophagy-related protein expression between ART-treated and untreated individuals underscore the intricate relationship between vitamin D₃ levels, ART exposure, and autophagic regulation. This is a preliminary exploration of the effects of vitamin D₃ on autophagy in PWH. Further studies are needed to deepen and explore the interplay between vitamin D₃ and autophagy in greater depth. A better understanding of these mechanisms could help to develop novel therapeutic strategies aimed at mitigating immune depletion and chronic inflammation, ultimately improving clinical outcomes for individuals living with HIV.
Collapse
Affiliation(s)
- Rita Casetti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - IRCCS, Rome, 00149, Italy
| | - Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - IRCCS, Rome, 00149, Italy
| | - Harpreet Kaur Lamsira
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Rome, 00131, Italy
| | - Carmela Pinnetti
- Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, 00149, Italy
| | - Valentina Mazzotta
- Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, 00149, Italy
| | - Serena Ciolfi
- Department of Science, University of Roma Tre, Rome, 00146, Italy
| | | | - Alessandra Amendola
- Laboratory of Virology and Biosafety Laboratories, National Institute for Infectious Diseases 'Lazzaro Spallanzani'- IRCCS, Rome, 00149, Italy
| | - Giuseppe Ippolito
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Rome, 00131, Italy
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'Lazzaro Spallanzani' - IRCCS, Rome, 00149, Italy
- Department of Biology, University of Rome 'Tor Vergata', Rome, 00133, Italy
| | - Roberta Nardacci
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, Rome, 00131, Italy.
| |
Collapse
|
10
|
Kowalski S, Wityk P, Raczak-Gutknecht J, Olszewska A, Żmijewski M, Kocić I. The imidazoline I 2 receptor agonist 2-BFI enhances cytotoxic activity of hydroxychloroquine by modulating oxidative stress, energy-related metabolism and autophagic influx in human colorectal adenocarcinoma cell lines. Eur J Pharmacol 2025; 996:177590. [PMID: 40185322 DOI: 10.1016/j.ejphar.2025.177590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Recently, interest in imidazoline receptors (IRs) has been increasing. Over the years, a growing number of studies have highlighted the therapeutic potential of ligands targeting these receptors, however, the potential role of imidazoline I2 receptor agonists in cancer treatment has not been thoroughly investigated. Colorectal cancer (CRC) is among the most prevalent and lethal forms of cancer worldwide. The complexity of CRC necessitates individualized approaches. One promising area of research within CRC therapy is the regulation of autophagy. Recent studies have explored the relationship between autophagy and cancer progression, revealing that autophagy modulation could be a potential strategy for CRC treatment. However, the mechanisms underlying autophagy regulation remain poorly understood. This study aimed to evaluate the effect of the imidazoline I2 receptor agonist, namely 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI), on colorectal cancer cells, HT-29 and HCT-116 cell lines, particularly its impact when co-incubated with the autophagy inhibitor, hydroxychloroquine (HCQ). The results showed that 2-BFI synergistically increased the cytotoxic effect of HCQ by inducing oxidative stress and apoptosis. Furthermore, our investigation indicated impairment autophagic influx in colon cancer cells treated by 2-BFI. The comprehensive metabolomic analysis revealed significant alterations in key metabolic pathways including MAO activity, oxidative stress responses, energy-related metabolites and amino acids metabolism. Altogether, these findings demonstrate potential a new therapeutic strategy based on autophagy regulation and the selective induction of oxidative stress in colorectal cancer cells. Moreover, this study provides a foundation for further investigation into the therapeutic potential of imidazoline receptor agonists in cancer therapy.
Collapse
Affiliation(s)
- Szymon Kowalski
- Department of Pharmacology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - Paweł Wityk
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland; Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, Gdansk, Poland; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joanna Raczak-Gutknecht
- Department of Biopharmaceutics and Pharmacodynamics, Faculty of Pharmacy, Medical University of Gdańsk, Gdansk, Poland
| | - Anna Olszewska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Michał Żmijewski
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ivan Kocić
- Department of Pharmacology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
11
|
Cheung YWS, Nam SE, Fairlie GMJ, Scheu K, Bui JM, Shariati HR, Gsponer J, Yip CK. Structure of the human autophagy factor EPG5 and the molecular basis of its conserved mode of interaction with Atg8-family proteins. Autophagy 2025; 21:1173-1191. [PMID: 39809444 PMCID: PMC12087653 DOI: 10.1080/15548627.2024.2447213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
The multi-step macroautophagy/autophagy process ends with the cargo-laden autophagosome fusing with the lysosome to deliver the materials to be degraded. The metazoan-specific autophagy factor EPG5 plays a crucial role in this step by enforcing fusion specificity and preventing mistargeting. How EPG5 exerts its critical function and how its deficiency leads to diverse phenotypes of the rare multi-system disorder Vici syndrome are not fully understood. Here, we report the first structure of human EPG5 (HsEPG5) determined by cryo-EM and AlphaFold2 modeling. Our structure revealed that HsEPG5 is constructed from helical bundles analogous to tethering factors in membrane trafficking pathways but contains a unique protruding thumb domain positioned adjacent to the atypical tandem LIR motifs involved in interaction with the GABARAP subfamily of Atg8-family proteins. Our NMR spectroscopic, molecular dynamics simulations and AlphaFold modeling studies showed that the HsEPG5 tandem LIR motifs only bind the canonical LIR docking site (LDS) on GABARAP without engaging in multivalent interaction. Our co-immunoprecipitation analysis further indicated that full-length HsEPG5-GABARAP interaction is mediated primarily by LIR1. Finally, our biochemical affinity isolation, X-ray crystallographic analysis, affinity measurement, and AlphaFold modeling demonstrated that this mode of binding is observed between Caenorhabditis elegans EPG-5 and its Atg8-family proteins LGG-1 and LGG-2. Collectively our work generated novel insights into the structural properties of EPG5 and how it potentially engages with the autophagosome to confer fusion specificity.ABBREVIATIONS: ATG: autophagy related; CSP: chemical shift perturbation; eGFP: enhanced green fluoresent protein; EM: electron microscopy; EPG5: ectopic P-granules 5 autophagy tethering factor; GST: glutathione S-transferase; HP: hydrophobic pocket; HSQC: heteronuclear single-quantum correlation; ITC: isothermal titration calorimetry; LDS: LC3 docking site; LIR: LC3-interacting region; MD: molecular dynamics; NMR: nuclear magnetic resonance; TEV: tobacco etch virus.
Collapse
Affiliation(s)
- Yiu Wing Sunny Cheung
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Sung-Eun Nam
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Gage M. J. Fairlie
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Karlton Scheu
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Jennifer M. Bui
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Hannah R. Shariati
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Jörg Gsponer
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Calvin K. Yip
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Gupta P, Sakthivel S, Jiemin N, Arul K, Tikno K, Park SH, Wu Y, Wang L, Bay BH, Ho L, Giguere V, Ghosh S, McDonnell DP, Yen PM, Singh BK. ESRRA (estrogen related receptor, alpha) induces ribosomal protein RPLP1-mediated adaptive hepatic translation during prolonged starvation. Autophagy 2025; 21:1283-1297. [PMID: 39936615 PMCID: PMC12087656 DOI: 10.1080/15548627.2025.2465183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, ESRRA (estrogen related receptor, alpha) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating Rplp1 (ribosomal protein lateral stalk subunit P1) gene expression. Overexpression or siRNA knockdown of Esrra in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome and macroautophagy/autophagy protein translation, and autophagy activity. Remarkably, we have found that ESRRA had dual functions by not only regulating transcription but also controlling adaptive translation via the ESRRA-RPLP1-lysosome-autophagy pathway during prolonged starvation.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Karine Gauthier
- Département de Biologie, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, Lyon, Cedex, France
| | - Reddemma Sandireddy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Priyanka Gupta
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Suganya Sakthivel
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Nah Jiemin
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Kabilesh Arul
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Keziah Tikno
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - Lena Ho
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, Canada
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| |
Collapse
|
13
|
Liu D, Weng S, Fu C, Guo R, Chen M, Shi B, Weng J. Autophagy in Acute Lung Injury. Cell Biochem Biophys 2025; 83:1415-1425. [PMID: 39527232 DOI: 10.1007/s12013-024-01604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is a critical condition marked by rapid-onset respiratory failure due to extensive inflammation and increased pulmonary vascular permeability, often progressing to acute respiratory distress syndrome (ARDS) with high mortality. Autophagy, a cellular degradation process essential for removing damaged organelles and proteins, plays a crucial role in regulating lung injury and repair. This review examines the protective role of autophagy in maintaining cellular function and reducing inflammation and oxidative stress in ALI. It underscores the necessity of precise regulation to fully harness the therapeutic potential of autophagy in this context. We summarize the mechanisms by which autophagy influences lung injury and repair, discuss the interplay between autophagy and apoptosis, and examine potential therapeutic strategies, including autophagy inducers, targeted autophagy signaling pathways, antioxidants, anti-inflammatory drugs, gene editing, and stem cell therapy. Understanding the role of autophagy in ALI could lead to novel interventions for improving patient outcomes and reducing mortality rates associated with this severe condition.
Collapse
Affiliation(s)
- Danjuan Liu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Shuoyun Weng
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Chunjin Fu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Rongjie Guo
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Min Chen
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Bingbing Shi
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Junting Weng
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China.
| |
Collapse
|
14
|
Zhu G, Zuo Q, Liu S, Zheng P, Zhang Y, Zhang X, Rollins JA, Liu J, Pan H. A FOX transcription factor phosphorylated for regulation of autophagy facilitates fruiting body development in Sclerotinia sclerotiorum. THE NEW PHYTOLOGIST 2025; 246:2683-2701. [PMID: 40248859 DOI: 10.1111/nph.70151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025]
Abstract
Autophagy is a recycling process by which eukaryotic cells degrade their own components, and the fruiting body (sexual structure) is a necessary structure for some plant pathogenic fungi to start the infection cycle. However, the transcriptional regulation of plant pathogenic fungal autophagy and autophagy regulating sexual reproduction remains elusive. Here, we provide the report linking autophagy transcription and fruiting body development in phytopathogenic fungi. The forkhead box transcription factor (FOX TF) SsFoxE2 in Sclerotinia sclerotiorum (Ss) binds to the promoters of ATG genes, thus promoting their transcription. SsFoxE2 is phosphorylated by AMP-activated protein kinase (AMPK) SsSnf1, and the phosphorylated SsFoxE2 interacts with (translationally controlled tumor protein) SsTctp1, leading to enhanced stability and ATG transcription activity of SsFoxE2. Importantly, the regulation of autophagy by SsFoxE2 affects the balance of the ubiquitination system and the early development of the fruiting body, which directly determines the occurrence and prevalence of plant disease. Furthermore, transcriptional binding of FOX TF to ATG gene promoters is conserved in phytopathogenic fungi. Taken together, our results bring new insights into pathogen initiation in phytopathogenic fungi and connect it to other autophagy-regulated processes in plant pathogens.
Collapse
Affiliation(s)
- Genglin Zhu
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Qi Zuo
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Sirui Liu
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Peiyi Zheng
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Yanhua Zhang
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Xianghui Zhang
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Jeffrey A Rollins
- Department of Plant Pathology, University of Florida, Gainesville, FL, 32611, USA
| | - Jinliang Liu
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| | - Hongyu Pan
- College of Plant Sciences, Jilin University, Changchun, 130062, China
| |
Collapse
|
15
|
Yamamoto T. Autophagic stagnation: a key mechanism in kidney disease progression linked to aging and obesity. Clin Exp Nephrol 2025; 29:711-719. [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] [MESH Headings] [Track Full Text] [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
|
16
|
Guo Z, He L, Wang W, Tian S, Lin R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett 2025; 619:217643. [PMID: 40112906 DOI: 10.1016/j.canlet.2025.217643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Fucosyltransferase 2 (FUT2) is an enzyme that adds fucose to proteins or lipids via α-1,2-fucosylation in the intestinal mucosa. While FUT2 deficiency is linked to increased susceptibility to inflammatory bowel disease (IBD), its role in colorectal cancer (CRC) is unclear, and the molecular mechanisms involved remain largely unknown. We established an azoxymethane (AOM) and dextran sulfate sodium (DSS) model to induce CRC. FUT2 expression was assessed in human CRC tissues, AOM/DSS-induced mouse models, and CRC cell lines using qRT-PCR, western blotting, and UEA-I staining. FUT2 knockout (FUT2△IEC) mice were treated with AOM/DSS, and FUT2-overexpressing CRC cells were created to evaluate the effects of FUT2 on apoptosis in both in vitro and in vivo settings through Western blot analyses and functional assays. N-glycoproteomics, UEA-I chromatography, and co-immunoprecipitation were utilized to identify regulatory mechanisms and target fucosylated proteins. FUT2 expression and α-1,2-fucosylation were significantly decreased in CRC. FUT2 deficiency worsened AOM/DSS-induced CRC and reduced tumor apoptosis, while FUT2 overexpression induced apoptosis and inhibited proliferation in CRC cells and xenografts. Mechanistically, FUT2 appears to suppress autophagy by impairing lysosomal function and directly targeting and fucosylating LAMP1, contributing to lysosomal dysfunction. Our study reveals a fucosylation-dependent antitumor mechanism of FUT2 in CRC, suggesting potential therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingnan He
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New Area, Shanghai, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Tian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Smith SG, Bowness R, Cliff JM. Host-directed therapy in diabetes and tuberculosis comorbidity toward global tuberculosis elimination. Int J Infect Dis 2025; 155:107877. [PMID: 40068707 DOI: 10.1016/j.ijid.2025.107877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/04/2025] Open
Abstract
Host-directed therapy could potentially revolutionize tuberculosis control as an adjunct to traditional antibiotics for the treatment of tuberculosis disease and as a strategy to prevent disease progression following Mycobacterium tuberculosis infection. The growing type 2 diabetes pandemic is hampering tuberculosis control worldwide, as people with diabetes have an increased risk of developing tuberculosis disease as well as worse treatment outcomes. Pulmonary tuberculosis is characterized by an inflammatory response that can cause alveolar tissue destruction and cavitation, and this inflammation is exacerbated in people with tuberculosis-diabetes comorbidity. Thus, the reduction of the inflammatory response is a key goal of host-directed therapy to dampen immunopathology, but it is vital that the inflammatory response is not suppressed too much, or the immune system will not be able to react to M. tuberculosis and mycobacterial replication will intensify. Furthermore, the type I interferon response and host cell metabolism are further dysregulated in tuberculosis-diabetes comorbidity, likely contributing to poor treatment outcomes. Achieving the right balance in terms of modulating the inflammatory and immune responses, both quantitatively and temporally, is more complex in tuberculosis-diabetes comorbidity, and this population should be included specifically in clinical trials of new regimens. In this regard, mathematical modeling has a key role in elucidating which biologic pathways should be targeted in different people. Host-directed therapy for people with tuberculosis-diabetes comorbidity will reduce immunopathology and post-tuberculosis lung disease, as well as boost microbiologic cure and treatment outcomes, and thus help in the fight toward global tuberculosis elimination.
Collapse
Affiliation(s)
- Steven G Smith
- Centre for Inflammation Research and Translational Medicine, Department of Biosciences, Brunel University of London, London, United Kingdom
| | - Ruth Bowness
- Centre for Mathematical Biology, Department of Mathematical Sciences, University of Bath, Bath, United Kingdom
| | - Jacqueline M Cliff
- Centre for Inflammation Research and Translational Medicine, Department of Biosciences, Brunel University of London, London, United Kingdom.
| |
Collapse
|
18
|
Kiani P, Khodadadi ES, Nikdasti A, Yarahmadi S, Gheibi M, Yousefi Z, Ehtiati S, Yahyazadeh S, Shafiee SM, Taghizadeh M, Igder S, Khatami SH, Karima S, Vakili O, Pourfarzam M. Autophagy and the peroxisome proliferator-activated receptor signaling pathway: A molecular ballet in lipid metabolism and homeostasis. Mol Cell Biochem 2025; 480:3477-3499. [PMID: 39891864 DOI: 10.1007/s11010-025-05207-0] [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: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
Lipids, which are indispensable for cellular architecture and energy storage, predominantly consist of triglycerides (TGs), phospholipids, cholesterol, and their derivatives. These hydrophobic entities are housed within dynamic lipid droplets (LDs), which expand and contract in response to nutrient availability. Historically perceived as a cellular waste disposal mechanism, autophagy has now been recognized as a crucial regulator of metabolism. Within this framework, lipophagy, the selective degradation of LDs, plays a fundamental role in maintaining lipid homeostasis. Dysregulated lipid metabolism and autophagy are frequently associated with metabolic disorders such as obesity and atherosclerosis. In this context, peroxisome proliferator-activated receptors (PPARs), particularly PPAR-γ, serve as intracellular lipid sensors and master regulators of gene expression. Their regulatory influence extends to both autophagy and lipid metabolism, indicating a complex interplay between these processes. This review explores the hypothesis that PPARs may directly modulate autophagy within the realm of lipid metabolism, thereby contributing to the pathogenesis of metabolic diseases. By elucidating the underlying molecular mechanisms, we aim to provide a comprehensive understanding of the intricate regulatory network that connects PPARs, autophagy, and lipid homeostasis. The crosstalk between PPARs and other signaling pathways underscores the complexity of their regulatory functions and the potential for therapeutic interventions targeting these pathways. The intricate relationships among PPARs, autophagy, and lipid metabolism represent a pivotal area of research with significant implications for understanding and treating metabolic disorders.
Collapse
Affiliation(s)
- Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elaheh Sadat Khodadadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122, Padova, Italy
| | - Ali Nikdasti
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Sahar Yarahmadi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mobina Gheibi
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeynab Yousefi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Motahareh Taghizadeh
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
19
|
Goletti D, Matteelli A, Cliff JM, Meintjes G, Graham S, Esmail H, Shan Lee S. World TB Day 2025 Theme "Yes! We Can End TB: Commit, Invest, Deliver" can be made a reality through concerted global efforts to advance diagnosis, treatment and research of tuberculosis infection and disease. Int J Infect Dis 2025; 155:107892. [PMID: 40107343 DOI: 10.1016/j.ijid.2025.107892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Affiliation(s)
- Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research National Institute for Infectious Diseases L. Spallanzani-IRCCS, Roma, Italy.
| | - Alberto Matteelli
- Clinic of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Jacqueline M Cliff
- Centre for Inflammation Research and Translational Medicine, Department of Biosciences, Brunel University of London, United Kingdom
| | - Graeme Meintjes
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Medicine and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Steve Graham
- Melbourne Children's Global Health, University of Melbourne Department of Paediatrics and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - Hanif Esmail
- WHO Collaborating Centre for Tuberculosis Research and Innovation; Centre for Global TB Research, Institute for Global Health; MRC Clinical Trials Unit, University College London, United Kingdom; Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine and Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Shui Shan Lee
- S.H. Ho Research Centre for Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China; International Society for Infectious Diseases
| |
Collapse
|
20
|
Huang C, Luo Y, Liu Y, Liu J, Chen Y, Zeng B, Liao X, Liu Y, Wang X. DNA hypermethylation-induced suppression of ALKBH5 is required for folic acid to alleviate hepatic lipid deposition by enhancing autophagy in an ATG12-dependent manner. J Nutr Biochem 2025; 140:109870. [PMID: 39993647 DOI: 10.1016/j.jnutbio.2025.109870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/08/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) occurs when too much fat builds up in the liver. As a growing worldwide epidemic, NAFLD is strongly linked with multiple metabolic diseases including obesity, insulin resistance, and dyslipidemia. However, very few effective treatments are currently available. Folate, an essential B-group vitamin with important biological functions including DNA and RNA methylation regulation, has been shown to have a protective effect against NAFLD with its underlying mechanism remains largely unclear. Here, we show that administration of folic acid significantly improves glucose tolerance, insulin sensitivity, and dyslipidemia in high-fat diet (HFD) fed mice. Moreover, folic acid treatment significantly inhibits lipid deposition in hepatocytes both in vivo and in vitro. Mechanically, folic acid reduces the expression of m6A demethylase AlkB homolog 5 (ALKHB5) via promoter DNA hypermethylation. Decreased ALKBH5 causes increased m6A modification and increased expression of ATG12 in a demethylase activity-dependent manner, thereby promoting autophagy and preventing hepatic steatosis. Inhibition of ATG12 induced by overexpression of ALKBH5 could impair autophagy and the inhibitory effect of folic acid on lipid accumulation in hepatocytes. Together, these findings provide novel insights into understanding the protective role of folic acid in the treatment of NAFLD and suggest that folic acid may be a potential agent for combating NAFLD.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Botao Zeng
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xing Liao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, Zhejiang, PR China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang, PR China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang, PR China.
| |
Collapse
|
21
|
Ji P, Li Q, Zhang Y, Jin J, Zhang Y, Yuan Z, Shen G, Cao Q, Wu Y, Wang P, Liu W. The role of RAB12 in inhibiting osteogenic differentiation and driving metabolic dysregulation in osteoporosis. Life Sci 2025; 370:123590. [PMID: 40147529 DOI: 10.1016/j.lfs.2025.123590] [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: 12/11/2024] [Revised: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
AIMS The osteogenic differentiation of mesenchymal stem cells (MSCs) is crucial in osteoporosis, and the metabolic level of the bone microenvironment directly affects metabolic dysregulation in postmenopausal women. RAB12 is a member of the small GTPase Rab family proteins, known to play an important role in autophagy. However, the role of RAB12 in the osteogenic differentiation of osteoporotic hMSCs remains unclear. MATERIALS AND METHOD Immunohistochemical staining was used to validate the high expression of RAB12 in aged osteoporotic mouse models and ovariectomized (OVX) mouse models. Co-immunoprecipitation (Co-IP) and LC-MS/MS were employed to explore downstream proteins that may interact with RAB12. Adenovirus containing RAB12 siRNA sequences was injected into the tail vein of OVX osteoporotic mice to analyze the impact of the RAB12/PCBP1/GLUT1 axis on MSC osteogenic differentiation. KEY FINDINGS We found that RAB12 expression is upregulated in elderly osteoporotic patients and in osteoporotic mouse models. RAB12 negatively regulates the osteogenic differentiation of hMSCs both in vivo and in vitro. RAB12 interacts with the PCBP1 protein, affecting its autophagic degradation when its expression levels change. RAB12 regulates the transcriptional level of GLUT1 by influencing the autophagic degradation of PCBP1, thereby affecting MSC's regulation of glucose uptake, which in turn impacts MSC osteogenic differentiation and metabolic changes. SIGNIFICANCE RAB12 negatively regulates osteogenic differentiation through the PCBP1/GLUT1 axis, affecting glucose metabolism levels in the bone microenvironment. RAB12 may serve as a potential target for the treatment of osteoporosis and postmenopausal metabolic dysregulation.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Quanfeng Li
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Yunhui Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Jiahao Jin
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Yibin Zhang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Zihao Yuan
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Guozhen Shen
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China
| | - Qian Cao
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China; Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China
| | - Yanfeng Wu
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China; Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China.
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, PR China; Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, PR China.
| |
Collapse
|
22
|
Costa RM, Bruder-Nascimento A, Alves JV, Awata WMC, Singh S, Rodrigues D, Bruder-Nascimento T, Tostes RC. Beclin-1-dependent autophagy protects perivascular adipose tissue function from hyperaldosteronism effects. Am J Physiol Heart Circ Physiol 2025; 328:H1253-H1266. [PMID: 40327449 DOI: 10.1152/ajpheart.00829.2024] [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: 11/27/2024] [Revised: 12/13/2024] [Accepted: 04/22/2025] [Indexed: 05/08/2025]
Abstract
Hyperaldosteronism (HA), characterized by excessive production of aldosterone (Aldo), contributes to cardiovascular damage and perivascular adipose tissue (PVAT) dysfunction. Previous studies have shown that Aldo can impair autophagy in various tissues. However, it remains unclear whether this impairment occurs specifically in PVAT and whether it involves disruption of autophagic flux through Beclin-1 (BCN1), a key regulator of autophagosome formation and maturation. We hypothesize that BCN1-dependent autophagy plays a protective role in PVAT by limiting inflammation and preserving its anticontractile function in the context of HA. Male and female C57BL/6J [wild type (WT)] and BCN1 knock-in mice, aged 10-12 wk, underwent 14-day aldosterone infusion (600 µg/kg/day) using an osmotic minipump. Vascular function was assessed in PVAT-intact thoracic aortae, and blood pressure was monitored via radiotelemetry. HA disrupted PVAT autophagic flux, leading to the accumulation of LC3II/I and p62 proteins and reduced BCN1 expression/activity. In WT mice, PVAT exhibited an anticontractile effect, which was abolished by HA. In contrast, BCN1-knock-in mice were protected from this loss of PVAT function. HA also induced oxidative stress and inflammation in PVAT, as evidenced by increased reactive oxygen species generation and elevated mRNA levels of TNF-α, IL-6, IL-1β, and IL-17. These proinflammatory and prooxidative changes were not observed in BCN1-knock-in mice, indicating preserved PVAT homeostasis. Furthermore, pharmacological induction of autophagy via spermidine and activation of BCN1 with TB peptide improved PVAT function in HA-treated WT mice. Finally, BCN1-knock-in mice exhibited partial protection against HA-induced hypertension, highlighting the systemic vascular benefits of enhanced autophagic flux. In summary, our findings demonstrate that the activation of autophagy provides protection against HA-induced PVAT inflammation, dysfunction, and hypertension. Consequently, the activation of BCN1 could serve as a pharmacological strategy to prevent the harmful cardiovascular effects associated with HA.NEW & NOTEWORTHY Elevated aldosterone levels, as seen in primary hyperaldosteronism, obesity, and hypertension, impair autophagic flux in perivascular adipose tissue (PVAT), leading to increased inflammation and loss of anticontractile function. The Beclin-1-dependent autophagic pathway plays a key role in maintaining PVAT homeostasis and vascular tone. Disrupted autophagy contributes to oxidative stress and hypertension. Activating this pathway may offer a novel therapeutic strategy to mitigate aldosterone's harmful vascular effects in hypertension by restoring PVAT function and vascular inflammation.
Collapse
Affiliation(s)
- Rafael M Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Institute of Health Sciences, Federal University of Jatai, Jatai, Brazil
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
| | - Ariane Bruder-Nascimento
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
| | - Juliano V Alves
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Wanessa M C Awata
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Shubhnita Singh
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Daniel Rodrigues
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Bruder-Nascimento
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
23
|
Seyyedin S, Nematollahi-Mahani SN. Bilateral asymmetrical variation of median artery in coexistence with bifid median nerve and variation in the origin and course of its palmar cutaneous branch: a case study with clinical implications. Anat Sci Int 2025; 100:366-369. [PMID: 39652155 DOI: 10.1007/s12565-024-00817-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/22/2024] [Indexed: 05/01/2025]
Abstract
The median artery typically regresses after two months of intrauterine life, although it may persist into adulthood in some individuals. The presence of a persistent median artery (PMA) may be associated with other anatomical variations including a bifid median nerve. In the present cadaveric study, we report a rare variation of bilateral asymmetry of PMA associated with the bifid median nerve, and unilateral variation of the origin and course of the palmar cutaneous branch of the median nerve (PCBMN) which to our knowledge, is the first study to report all these variations in an individual. Classical dissection of the upper limb was performed on a 45-year-old male cadaver. The cadaver was donated to the Department of Anatomy at Kerman University of Medical Sciences. Bilateral PMA was observed in both upper limbs. The PMA originated from the ulnar artery and contributed to the formation of an incomplete superficial palmar arch (SPA) on both sides; however, the branching pattern of these arteries was different between the right and left hands. Also, a bilateral high division of the median nerve was observed proximal to the carpal tunnel. We also encountered a very rare variation of PCBMN, in which it originated from the ulnar side of the median nerve, and passed beneath the flexor retinaculum of the left hand. Awareness of anatomical variations of the median nerve and also the presence of PMA is of utmost importance due to their implication in carpal tunnel syndrome and surgical complications.
Collapse
Affiliation(s)
- Sajad Seyyedin
- Department of Anatomical Sciences, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
24
|
Sugawara Y, Morinaga H, Chen J, Kitagawa Y, Ogata H, Karim A, Kikuchi M, Khan M, Yasuhara E, Goto T, Martyn JAJ, Yasuhara S. Mito-Kaede photoactivation and chase experiment for mitophagy: mitophagy flux response toward various stimulations. Biotechniques 2025:1-13. [PMID: 40449520 DOI: 10.1080/07366205.2025.2505357] [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: 12/24/2023] [Accepted: 05/09/2025] [Indexed: 06/03/2025] Open
Abstract
Mitophagy, a crucial mitochondrial quality control system for cellular stress adaptation, is a key focus in pathophysiology and drug discovery. Developing a simple and versatile mitophagy flux assay is vital for advancing our understanding of cellular responses. Addressing a gap in systematic methods, we employ the photoactivatable fluorescent protein mito-Kaede in C2C12 myocytes, demonstrating its remarkable versatility in quantifying mitophagy flux responses under various stimuli, including carbonyl cyanide m-chlorophenyl hydrazone (CCCP), TNF-α, lipopolysaccharide (LPS), and hypoxia. This study underscores the validity and distinctive advantages of the mito-Kaede assay through comparative analysis with conventional assays including Western blotting (WB), potentially providing valuable insights for both mitophagy flux analysis and drug development.
Collapse
Affiliation(s)
- Yoh Sugawara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Morinaga
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
- Department of Trauma and Critical Care Medicine, Kyorin University, Faculty of Medicine
| | - Jingyuan Chen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yoshinori Kitagawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Hiroki Ogata
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Asiya Karim
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Miu Kikuchi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Maryam Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Erica Yasuhara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Takahisa Goto
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Joseph A Jeevendra Martyn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| | - Shingo Yasuhara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children Boston, and Harvard Medical School, Boston, MS, USA
| |
Collapse
|
25
|
Anifowose SO, Oladejo MK, Salih AM, Almutairi LM, Almansour MI, Al-Dahmash B, Al Mosallam MS, Alanazi IO, Rady A. Dual Modulation of Autophagy and Apoptosis as Anticancer Mechanism of Action of Khaya grandiofoliola in Colon Carcinoma Cells. Int J Mol Sci 2025; 26:5247. [PMID: 40508060 PMCID: PMC12155393 DOI: 10.3390/ijms26115247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/26/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Khaya grandiofoliola (Kh) is a medicinal plant with therapeutic properties. Studies have reported on the general bioactivity and anticancer potentials of the plant, but no investigations have yet investigated its anticancer mechanism of action. This study presents the first examination of the anticancer mechanism of action of the methanolic extract of Kh, alongside phytochemical profiling of its anticancer constituents. We conducted in vitro investigations into the mechanism of action of Kh and performed bioactivity-guided fractionation, with subsequent identification of its anticancer phytochemicals using HPLC and GC-MS, respectively. Kh posed a potent antiproliferative effect against colon carcinoma cells and an antioxidant property at low microgram levels. Furthermore, the treatment of Kh in Caco-2 cells led to the accumulation of p62 puncta, indicating inhibition of autophagic flux degradation. Kh impacts microtubule, induced G1 arrest, and late apoptosis induction in Caco-2 cells. Phytochemicals belonging to sesquiterpene alcohols were found most abundant in the Kh bioactive fractions. The identified phytochemicals are potential inducers of apoptosis, autophagy flux inhibition, and G1-phase arrest. Our findings suggest that the anticancer property of Kh is mediated through the dual modulation of autophagy and apoptosis. Further studies are needed to isolate the active compounds responsible for these effects and further elucidate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Saheed O. Anifowose
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (S.O.A.)
| | - Musa K. Oladejo
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (S.O.A.)
| | - Abdalrhaman M. Salih
- Botany and Microbiology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Layali M. Almutairi
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia
| | - Mansour I. Almansour
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (S.O.A.)
| | - Badr Al-Dahmash
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (S.O.A.)
| | - Mobarak S. Al Mosallam
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Ibrahim O. Alanazi
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - Ahmed Rady
- Zoology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (S.O.A.)
| |
Collapse
|
26
|
Cai YY, Zhu XM, Noman M, Wang J, Hao ZN, Wang YL, Li L, Liu XH, Lu JP, Wang JY, Lin FC. Nuclear basket nucleoporin MoNup50 is essential for fungal development, pathogenicity, and autophagy in Magnaporthe oryzae. Cell Commun Signal 2025; 23:249. [PMID: 40442711 PMCID: PMC12121117 DOI: 10.1186/s12964-025-02219-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/26/2025] [Indexed: 06/02/2025] Open
Abstract
Autophagy is crucial for appressorium development and host invasion by phytopathogenic fungi, including Magnaporthe oryzae. During appressorium maturation, many organelles, such as nuclei, in the conidia need to be degraded through autophagy to be recycled in appressorium. However, the interplay between autophagy and nuclear membrane systems remains poorly understood. In this study, we functionally characterized MoNup50, a nuclear pore-associated protein. Despite sharing limited sequence identity with human and yeast Nup proteins, MoNup50 contains conserved domains typical of nuclear pore complex proteins. Observation under fluorescence microscopy revealed that MoNup50 localizes at the nuclear membrane in M. oryzae. Deletion of MoNUP50 resulted in reduced hyphal growth, spore production, appressorium formation, and pathogenicity, while increasing sensitivity to osmotic stress and cell wall disruption. Notably, MoNup50 interacts with the key autophagy protein MoAtg7, which regulates MoAtg8-PE synthesis during autophagy. Moreover, MoNUP50 deletion led to elevated autophagy levels and increased phosphorylation of the MAPKs Osm1 and Mps1. These findings suggest that MoNup50 is involved in appressorium morphogenesis and pathogenicity by modulating autophagy and MAPK pathways, highlighting the critical role of nuclear pore proteins in M. oryzae pathogenicity and their potential cross-talk with autophagic and MAPK signaling.
Collapse
Affiliation(s)
- Ying-Ying Cai
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Xue-Ming Zhu
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Muhammad Noman
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Jing Wang
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Zhong-Na Hao
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yan-Li Wang
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Lin Li
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Xiao-Hong Liu
- State Key Laboratory for Quality and Safety of Agro-Products, Key Laboratory of Agricultural Microbiomics of Zhejiang Province, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Jian-Ping Lu
- College of Life Science, Zhejiang University, Hangzhou, 310058, China
| | - Jiao-Yu Wang
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Fu-Cheng Lin
- State Key Laboratory for Quality and Safety of Agro-products, Key Laboratory of Agricultural Microbiome (MARA), Key Laboratory of Agricultural Microbiomes of Zhejiang Province, Key Laboratory of Biotechnology in Plant Protection of MARA, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
- State Key Laboratory for Quality and Safety of Agro-Products, Key Laboratory of Agricultural Microbiomics of Zhejiang Province, Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
27
|
Zhang H, Liang H, Fan L, Zhu X, Ji P, Su Y, Li W, Li W. Ginsenoside Rg1 attenuates T2DM-induced renal damage and fibrosis by inhibiting TRPC6-ChREBP-TXNIP signaling. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119863. [PMID: 40311716 DOI: 10.1016/j.jep.2025.119863] [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: 02/25/2025] [Revised: 04/12/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a traditional Chinese medicine, ginseng has many benefits, including regulating blood sugar, blood pressure and so on. Ginsenoside Rg1 is the main active component of ginseng and has been found to significantly improve renal pathological injury in type 2 diabetes mellitus (T2DM) mice. However, the effects and mechanisms of Rg1 in attenuating T2DM are not fully understood. AIM OF THE STUDY This study aims to investigate the role of Rg1 in the treatment of renal damage and fibrosis induced by T2DM and its molecular mechanism. MATERIALS AND METHODS T2DM models were constructed on mice and cells respectively and were administered with corresponding drugs. SA-β-Gal and Oil Red O were used to observe cell senescence and lipid droplet deposition; H&E and PAS were used to observe pathological changes in the kidney; masson and sirius red were used to evaluate the level of renal fibrosis. Immunohistochemistry, immunofluorescence and Western blotting were performed to analyze the relevant indexes which resulted in the detection of ROS levels in vitro and in vivo. Calcium imaging was used to test the level of [Ca2+]i. RESULTS Rg1 and Trpc6 knockout could significantly improve kidney dysfunction, attenuate renal injury and fibrosis and also decrease the expression levels of TRPC6, CaN, TXNIP, ChREBP, p-ASK1 and NLRP3 inflammasome. Meanwhile, Rg1 and Trpc6 knockout significantly inhibited mitochondrial damage and apoptosis protein release. Additionally, Rg1 treatment has been shown to markedly reduce lipid deposition and ROS accumulation in T2DM, while Trpc6 knockout exhibited no effect on these parameters. CONCLUSION Rg1 treatment can inhibit the TRPC6-ChREBP-TXNIP pathway, thereby improving chronic T2DM-induced renal injury and fibrosis.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Haoyu Liang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Lei Fan
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xing Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Pengmin Ji
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Weiping Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weizu Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
28
|
Barros D, Ferreira BH, Garcia-Gonzalez P, Carbone F, Luka M, Leite-Pinheiro F, Machado MD, Nikolaou T, Pilotti A, Goguet E, Antas P, Mendes A, Zhang L, Cresci M, Galliot L, Gigan JP, Reverendo M, Su B, Narita M, Paton AW, Paton JC, Rocchi S, Rieux-Laucat F, Argüello RJ, Nal B, Liang Y, Ménager M, Gatti E, Almeida CR, Pierre P. Induction of the ISR by AB5 subtilase cytotoxin drives type-I IFN expression in pDCs via STING activation. Proc Natl Acad Sci U S A 2025; 122:e2421258122. [PMID: 40388626 DOI: 10.1073/pnas.2421258122] [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: 10/15/2024] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
We demonstrate that exposure to the AB5 subtilase cytotoxin (SubAB) induces the unfolded protein response (UPR) in human peripheral blood mononuclear cells, concomitant with a proinflammatory response across distinct cell subsets. Notably, SubAB selectively induces type-I interferon (IFN) expression in plasmacytoid dendritic cells, acting synergistically with Toll-like receptor 7 stimulation. The induction of type-I IFN in response to SubAB relies on stimulator of interferon genes (STING) activation, coupled with protein synthesis inhibition mediated by protein kinase R-like endoplasmic reticulum kinase (PERK) and phosphorylation of the eukaryotic translation initiation factor 2 subunit-alpha. By impeding mRNA translation through the integrated stress response, SubAB precipitates the downregulation of the negative innate signaling feedback regulator Tax1-binding protein 1. This downregulation is necessary to unleash TANK-binding kinase 1 signaling associated with STING activation. These findings shed light on how UPR-inducing conditions may regulate the immune system during infection or pathogenesis.
Collapse
Affiliation(s)
- Daniela Barros
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Beatriz H Ferreira
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Paulina Garcia-Gonzalez
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Francesco Carbone
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Marine Luka
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Fátima Leite-Pinheiro
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Mariana D Machado
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Theopisti Nikolaou
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Angelo Pilotti
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Eliot Goguet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Paulo Antas
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Andreia Mendes
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Lichen Zhang
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Marina Cresci
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Lou Galliot
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Julien P Gigan
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Marisa Reverendo
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Bing Su
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Miwako Narita
- Laboratory of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata 951-8518, Japan
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Frédéric Rieux-Laucat
- Université de Paris Cité, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris 75015, France
| | - Rafael J Argüello
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Béatrice Nal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Yinming Liang
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Mickaël Ménager
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Evelina Gatti
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Catarina R Almeida
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Philippe Pierre
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| |
Collapse
|
29
|
Lai WY, Chuang CW, Huang YC, Huang CJ. Therapeutic Potential of Plant-Derived Small Extracellular Vesicles in Sepsis: A Network Meta-analysis. Pharmacol Res 2025:107795. [PMID: 40414583 DOI: 10.1016/j.phrs.2025.107795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Sepsis is a life-threatening condition characterized by systemic inflammation and multi-organ dysfunction. Plant-derived small extracellular vesicles (sEVs) have emerged as promising therapeutic agents due to their antioxidant, anti-inflammatory, and immunomodulatory properties. This study conducted a network meta-analysis to identify the most effective plant-derived sEVs for reducing sepsis-induced inflammation and oxidative stress. The analysis included 13 studies evaluating 10 plant-derived sEVs in sepsis-mimicking conditions, with primary outcomes focused on cytokine levels and reactive oxygen species (ROS) production in vitro and in vivo. Secondary outcomes included nuclear factor erythroid 2-related factor 2 (Nrf2) expression and cell viability. The study protocol was registered with PROSPERO (CRD420251011005). Ginger-derived sEVs were identified as the most effective, significantly reducing pro-inflammatory cytokines (interleukin-6 and tumor necrosis factor-α), increasing the anti-inflammatory cytokine (interleukin-10), and suppressing ROS production. They also enhanced Nrf2 expression and improved cell viability, highlighting their role in antioxidant defense and cytoprotection. In conclusion, ginger-derived sEVs are the most effective plant-derived sEVs for mitigating sepsis-induced inflammation and oxidation in both in vitro and in vivo sepsis-mimicking models.
Collapse
Affiliation(s)
- Wen-Yi Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wei Chuang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Jen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
30
|
Ranjana M, Shrilaxmi MS, Nag P, Mitra PK, Sunil D, Dastidar SG, Sudhakar YN, Vennapusa SR, Raju R, Tittonen I, Upadhya D. A versatile electrochemical, colorimetric, and visible light excitable turn-on fluorescent probe for stress-induced H 2S detection. Photochem Photobiol Sci 2025:10.1007/s43630-025-00739-2. [PMID: 40402359 DOI: 10.1007/s43630-025-00739-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Hydrogen sulfide (H2S) holds a distinct role in cell biology. Its level is intricately linked to the homeostasis of the biological environment, underscoring the significance of developing techniques capable of detecting H2S in biological systems. A single probe that offers versatility across different detection techniques opens opportunities for advancements in sensing H2S in various fields. A nitronaphthalimide derivative, NMO prepared using a simple synthetic protocol, has been studied as an electrochemical, colorimetric, and turn-on fluorescence probe for H2S. NMO displayed a detection limit of 9.95 mM and 4.36 mM in the UV-visible and colorimetric studies, respectively, whereas the fluorometric and square wave techniques confirmed lower detection limits of 98.4 μM and 1.24 mM, correspondingly. Further, the real-time imaging of HEK293T cells using NMO during stress-induced autophagy is demonstrated.
Collapse
Affiliation(s)
- M Ranjana
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - M S Shrilaxmi
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Probal Nag
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, 695016, India
| | - Prajoy Kumar Mitra
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, 695016, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Somasish Ghosh Dastidar
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Y N Sudhakar
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sivaranjana Reddy Vennapusa
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, 695016, India
| | - Ramesh Raju
- Department of Electronics and Nanoengineering, Aalto University, P O Box 13500, 00076, Espoo, Finland
| | - Ilkka Tittonen
- Department of Electronics and Nanoengineering, Aalto University, P O Box 13500, 00076, Espoo, Finland
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
31
|
Ullern H, Schnur P, Boccara CN, Knævelsrud H. Rest, Repair, Repeat: The Complex Relationship of Autophagy and Sleep. J Mol Biol 2025:169227. [PMID: 40409707 DOI: 10.1016/j.jmb.2025.169227] [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: 12/11/2024] [Revised: 05/13/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025]
Abstract
Autophagy and sleep are two evolutionary conserved mechanisms across the animal kingdom. Autophagy is a pathway for the degradation of cytoplasmic material in the lysosome, playing important roles in the homeostasis and health of the organism. On the other hand, sleep is a homeostatically regulated state with numerous presumed essential roles, including the restoration of tissue and physiological functions, such as brain waste clearance via the activation of the glymphatic systems. Given that sleep and autophagy are crucial processes tightly linked to homeostasis and maintenance of good health, understanding how they interact is of great interest, especially as sleep quality decreases in our modern 24-hour societies. Autophagy represents a promising target for therapeutic interventions in this context. Here, we review the contrasted and complementary roles of autophagy and sleep in maintaining homeostasis. Specifically, we focus on recent evidence suggesting that sleep impairment may increase autophagy, while autophagosome levels may modulate the amount of sleep. We discuss outstanding questions at the intersection of these two fields, highlighting methodological shortcomings in the current literature. Overcoming these limitations will be instrumental to design new experiments with the aim of answering one of the greatest mysteries of our time - why do we sleep?
Collapse
Affiliation(s)
- Halvor Ullern
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Paulina Schnur
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Norwegian Centre for Molecular Biosciences and Medicine (NCMBM), University of Oslo, Oslo, Norway
| | - Charlotte N Boccara
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Norwegian Centre for Molecular Biosciences and Medicine (NCMBM), University of Oslo, Oslo, Norway; Department of Neurology, Clinical Neuroscience, Oslo University Hospital (OUS), Norway.
| | - Helene Knævelsrud
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Norway.
| |
Collapse
|
32
|
Serot C, Scarcelli V, Pouget A, Largeau C, Sagot A, El-Hachami K, Dupuy D, Culetto E, Lefebvre C, Legouis R. Reticulon-dependent ER-phagy mediates adaptation to heat stress in C. elegans. Curr Biol 2025; 35:2365-2378.e7. [PMID: 40328253 DOI: 10.1016/j.cub.2025.04.028] [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: 07/26/2024] [Revised: 03/06/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025]
Abstract
The selective degradation of endoplasmic reticulum (ER) by autophagy, named ER-phagy, promotes the recovery of ER homeostasis after stress. Depending on the ER stress, different types of ER-phagy involve various selective autophagy receptors. In this study, we report a macroER-phagy induced by the fragmentation of tubular ER in response to acute heat stress. We identified a novel ER-phagy receptor encoded by the reticulon long isoform RET-1d. RET-1d is mainly expressed in the nervous system and the epidermis and colocalizes with the ubiquitin-like autophagy protein LGG-1/GABARAP during heat-stress-induced autophagy. Two LC3-interacting region (LIR) motifs in the long intrinsically disordered region of RET-1d mediate its interaction with the LGG-1 protein. The specific depletion of the RET-1d isoform or the mutations of the LIRs resulted in a defective ER-phagy and a decrease in the capacity of animals to adapt to heat stress. Our data revealed a RET-1d- and LGG-1-dependent ER-phagy mechanism that takes place in neurons and epidermis and participates in the adaptation of C. elegans to heat stress.
Collapse
Affiliation(s)
- Claudia Serot
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; Department of R&I in Monogastric Animal Nutrition, European Laboratory of Innovation Science & Expertise (ELISE), Adisseo France S.A.S., Saint Fons 69190, France
| | - Vincent Scarcelli
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Alexandre Pouget
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; INSERM U1280, Gif-sur-Yvette cedex 91198, France
| | - Céline Largeau
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; INSERM U1280, Gif-sur-Yvette cedex 91198, France
| | - Audrey Sagot
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; INSERM U1280, Gif-sur-Yvette cedex 91198, France
| | - Kenza El-Hachami
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Denis Dupuy
- University of Bordeaux, INSERM U1212, CNRS UMR 5320, ARN: Régulation Naturelle et Artificielle (ARNA) Laboratory, Bordeaux F-33000, France
| | - Emmanuel Culetto
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; INSERM U1280, Gif-sur-Yvette cedex 91198, France
| | - Christophe Lefebvre
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; INSERM U1280, Gif-sur-Yvette cedex 91198, France.
| | - Renaud Legouis
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France; INSERM U1280, Gif-sur-Yvette cedex 91198, France.
| |
Collapse
|
33
|
Clark-Flores D, Vidal-Montiel A, Mondragón-Flores R, Valentín-Gómez E, Hernández-Rodríguez C, Juárez-Montiel M, Villa-Tanaca L. Vacuolar Proteases of Candida auris from Clades III and IV and Their Relationship with Autophagy. J Fungi (Basel) 2025; 11:388. [PMID: 40422722 DOI: 10.3390/jof11050388] [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: 04/01/2025] [Revised: 05/03/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025] Open
Abstract
Candida auris is a multidrug-resistant pathogen with a high mortality rate and widespread distribution. Additionally, it can persist on inert surfaces for extended periods, facilitating its transmissibility in hospital settings. Autophagy is a crucial cellular mechanism that enables fungal survival under adverse conditions. A fundamental part of this process is mediated by vacuolar proteases, which play an essential role in the degradation and recycling of cellular components. The present work explores the relationship between C. auris vacuolar peptidases and autophagy, aiming to establish a precedent for understanding the survival mechanisms of this emerging fungus. Thus, eight genes encoding putative vacuolar peptidases in the C. auris genomes were identified: PEP4, PRB1, PRC1, ATG42, CPS, LAP4, APE3, and DAP2. Analysis of the protein domains and their phylogenetic relationships suggests that these enzymes are orthologs of Saccharomyces cerevisiae vacuolar peptidases. Notably, both vacuolar protease gene expression and the proteolytic activity of cell-free extracts increased under nutritional stress and rapamycin. An increase in the expression of the ATG8 gene and the presence of autophagic bodies were also observed. These results suggest that proteases could play a role in yeast autophagy and survival during starvation conditions.
Collapse
Affiliation(s)
- Daniel Clark-Flores
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico
| | - Alvaro Vidal-Montiel
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico
| | - Ricardo Mondragón-Flores
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN No. 2508, Ciudad de México 07360, Mexico
| | - Eulogio Valentín-Gómez
- Departmento de Microbiología y Ecología, Universidad de Valencia, 46100 Valencia, Spain
- Severe Infection Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - César Hernández-Rodríguez
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico
| | - Margarita Juárez-Montiel
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico
| | - Lourdes Villa-Tanaca
- Laboratorio de Biología Molecular de Bacterias y Levaduras, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. de Carpio y Plan de Ayala. Col. Sto. Tomás, Ciudad de México 11340, Mexico
| |
Collapse
|
34
|
Du A, Feng S, Zhou X, Li Y, Lu S, Wu B. Effect of NiCl2 Intake Through Respiratory Tract on Antioxidant Capacity, Lung, and Trace Element Content in Mice. Biol Trace Elem Res 2025:10.1007/s12011-025-04630-0. [PMID: 40381093 DOI: 10.1007/s12011-025-04630-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/16/2025] [Indexed: 05/19/2025]
Abstract
This study examined the acute respiratory toxicity of NiCl2 in mice, focusing on oxidative stress, tissue damage, and trace element dysregulation. Forty male KM mice were allocated to a saline control group and three NiCl2 exposure groups (20, 60, 115 mg/kg; n = 10/group). Serum analysis assessed oxidative stress (MDA, GSH, SOD), liver (AST, ALT), kidney (Cr, BUN) function, and TP. Lung and tracheal tissues were examined for histopathological/ultrastructural pathological changes and apoptosis. Tissue levels of Ni, Zn, Cu, Fe, Ca, Mg, and Mn were measured using spectrophotometry. Results revealed dose-responsive elevations in serum AST, ALT, BUN, Cr, and MDA, accompanied by diminished GSH, TP, and T-SOD (P < 0.05). Nickel exposure caused tracheal pseudostratified columnar epithelium detachment, alveolar structural wall thickening and widened septa, capillary congestion, mitochondrial swelling in alveolar type-II cells, and increased pulmonary apoptosis (P < 0.05). Ni accumulated predominantly in the liver, lung, and kidney, with concurrent Zn upregulation and Cu/Fe depletion (P < 0.05), while Ca, Mg, and Mn levels remained stable. These findings demonstrate that acute NiCl2 inhalation induces oxidative stress, impairs liver/kidney function, and provokes pulmonary apoptosis and mitochondrial damage. Ni disrupted Cu/Zn/Fe homeostasis but exhibited negligible effects on Ca, Mg, or Mn metabolism.
Collapse
Affiliation(s)
- Aifei Du
- College of Life Science, China West Normal University, Nanchong, 637000, Sichuan, China
| | - Shaohua Feng
- College of Life Science, China West Normal University, Nanchong, 637000, Sichuan, China
| | - Xuan Zhou
- College of Life Science, China West Normal University, Nanchong, 637000, Sichuan, China
| | - Yiyin Li
- College of Life Science, China West Normal University, Nanchong, 637000, Sichuan, China
| | - Shangqing Lu
- College of Life Science, China West Normal University, Nanchong, 637000, Sichuan, China
| | - Bangyuan Wu
- College of Life Science, China West Normal University, Nanchong, 637000, Sichuan, China.
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), Nanchong, 637000, Sichuan, China.
- Nanchong Key Laboratory of Wildlife Nutrition Ecology and Disease Control, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
35
|
He F, Nichols RM, Agosto MA, Wensel TG. Roles of class III phosphatidylinositol 3-kinase, Vps34, in phagocytosis, autophagy, and endocytosis in retinal pigmented epithelium. iScience 2025; 28:112371. [PMID: 40330883 PMCID: PMC12052997 DOI: 10.1016/j.isci.2025.112371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/17/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Phosphatidylinositol-3-phosphate (PI(3)P) is important for multiple functions of retinal pigmented epithelial (RPE) cells, but its functions in RPE have not been studied. In RPE from mouse eyes and in cultured human RPE cells, PI(3)P-enriched membranes include endosomes, the trans-Golgi network, phagosomes, and autophagophores. Mouse RPE cells lacking activity of the PI-3 kinase, Vps34, lack detectable PI(3)P and die prematurely. Phagosomes containing rod discs accumulate, as do membrane aggregates positive for autophagosome markers. These autophagy-related membranes recruit LC3/Atg8 without Vps34, but phagosomes do not. Vps34 loss leads to accumulation of lysosomes which do not fuse with phagosomes or membranes with autophagy markers. Thus, Vps34-derived PI(3)P is not needed for initiation of phagocytosis or endocytosis, nor for formation of membranes containing autophagy markers. In contrast, Vps34 and PI(3)P are essential for intermediate and later stages, including membrane fusion with lysosomes.
Collapse
Affiliation(s)
- Feng He
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Ralph M. Nichols
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Melina A. Agosto
- Retina and Optic Nerve Research Laboratory, Department of Physiology and Biophysics, and Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Theodore G. Wensel
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
36
|
Hao Y, Fan X, Huang X, Li Z, Jing Z, Zhang G, Xu Y, Zhang N, Wei P. Recovery of Lysosomal Acidification and Autophagy Flux by Attapulgite Nanorods: Therapeutic Potential for Lysosomal Disorders. Biomolecules 2025; 15:728. [PMID: 40427621 PMCID: PMC12109497 DOI: 10.3390/biom15050728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Dysfunction of the lysosome and autophagy-lysosome pathway is closely associated with various diseases, such as neurodegenerative diseases, non-alcoholic fatty liver disease (NAFLD), etc. Additionally, chloroquine is a clinically widely used drug for treating malaria and autoimmune diseases, but long-term or high-dose administration may lead to significant toxic side effects. Attapulgite (ATT), a natural nanomaterial with excellent adsorption capacity and biocompatibility, herein demonstrated a novel biological function in regulating the lysosomal and autophagy-lysosome pathway. ATT could be effectively internalized into lysosome-related acidic compartments. Further study revealed that ATT could restore lysosomal pH, activate cathepsin D, alleviate autophagy blockage in chloroquine-treated cells, and reduce chloroquine-elicited cell death. In a cell model related to Huntington's disease, treatment with ATT reinforced the degradation of the mutant huntingtin proteins by increasing cathepsin D maturation and autophagy flux. ATT could also promote lipid droplet clearance in hepatocytes with palmitic acid-induced steatosis, reduce hepatic lipid accumulation, and improve fasting blood glucose in high-fat-diet-induced NAFLD mice. These findings establish ATT as a lysosomal modulator, providing a foundation for its therapeutic potential in mitigating the adverse effects associated with long-term chloroquine use, especially improving neurodegenerative and metabolic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Na Zhang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, China; (Y.H.); (X.F.); (X.H.); (Z.L.); (Z.J.); (G.Z.); (Y.X.)
| | - Pengfei Wei
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, China; (Y.H.); (X.F.); (X.H.); (Z.L.); (Z.J.); (G.Z.); (Y.X.)
| |
Collapse
|
37
|
Shi P, Tang B, Xie W, Li K, Guo D, Li Y, Yao Y, Cheng X, Xu C, Wang QK. LncRNA-induced lysosomal localization of NHE1 promotes increased lysosomal pH in macrophages leading to atherosclerosis. J Biol Chem 2025:110246. [PMID: 40383150 DOI: 10.1016/j.jbc.2025.110246] [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: 01/31/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
ANRIL, also referred to as CDKN2B-AS1, is a lncRNA gene implicated in the pathogenesis of multiple human diseases including atherosclerotic coronary artery disease, however, definitive in vivo evidence is lacking and the underlying molecular mechanism is largely unknown. In this study, we show that ANRIL overexpression causes atherosclerosis in vivo as transgenic mouse overexpression of full-length ANRIL (NR_003529) increases inflammation and aggravates atherosclerosis under ApoE-/- background (ApoE-/-ANRIL mice). Mechanistically, ANRIL reduces the expression of miR-181b-5p, which leads to increased TMEM106B expression. TMEM106B is significantly up-regulated in atherosclerotic lesions of both human CAD patients and ApoE-/-ANRIL mice. TMEM106B interacts and co-localizes with Na+-H+ exchanger NHE1, which results in mis-localization of NHE1 from cell membranes to lysosomal membranes, leading to increased lysosomal pH in macrophages. Large truncation and point mutation analyses define the critical amino acids for TMEM106B-NHE1 interaction and lysosomal pH regulation as F115 and F117 on TMEM106B and I537, C538, and G539 on NHE1. Topological analysis suggests that both N-terminus and C-terminus of NHE1 are located inside lysosomal lumen, and NHE1 is an important new proton efflux channel involved in raising lysosomal pH. A short TMEM106B peptide (YGRKKRRQRRR-L111A112V113F114F115L116F117) disrupting the TMEM106B-NHE1 interaction normalized lysosomal pH in macrophages with ANRIL overexpression. Our data demonstrate that ANRIL promotes atherosclerosis in vivo and identify the ANRIL/miR-181b-5p/TMEM106B-NHE1/lysosomal pH axis as the underlying molecular pathogenic mechanism for the chromosome 9p21.3 genetic locus for coronary artery disease.
Collapse
Affiliation(s)
- Pengcheng Shi
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Bo Tang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Wen Xie
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Ke Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Di Guo
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Yining Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Yufeng Yao
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College
| | - Chengqi Xu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Qing K Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology; Maternal and Child Health Hospital of Hubei Province, Women and Children's Hospital of Hubei Province, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
38
|
Yi X, Abas R, Raja Muhammad Rooshdi RAW, Yan J, Liu C, An J, Daut UN. Time-restricted feeding attenuated hypertension-induced cardiac remodeling by modulating autophagy levels in spontaneously hypertensive rats. Sci Rep 2025; 15:16973. [PMID: 40374761 PMCID: PMC12081920 DOI: 10.1038/s41598-025-01587-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
To investigate whether time-restricted feeding (TRF) can alleviate cardiac remodeling in spontaneously hypertensive rats (SHRs) by regulating autophagy levels. A 16-week TRF intervention was conducted on Wistar Kyoto (WKY) rats and SHRs, with dietary intake confined to the interval from 9:00 am to 5:00 pm each day. The study examined the impact of TRF on blood pressure (BP), cardiac morphology and function, and the expression levels of key proteins involved in autophagy and its associated signaling cascades. Transmission Electron Microscopy (TEM) was utilized to further evaluate autophagic changes in left ventricular (LV) tissues. TRF significantly mitigated systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean blood pressure (MBP) in SHRs. Additionally, TRF improved ejection fraction (EF) and diminished interventricular septal thickness at end-diastole (IVS-d). The study further revealed that TRF enhanced the expression of microtubule-associated protein-I light chain 3 (LC3-I), while reducing that of microtubule-associated protein-II light chain 3 (LC3-II). Moreover, TRF suppressed the expression levels of Beclin-1, phosphorylated phosphoinositide 3-kinase (p-PI3K), phosphorylated protein kinase B (p-AKT), and phosphorylated mechanistic target of rapamycin (p-mTOR) in the LV tissues. TEM analysis confirmed that TRF could inhibit autophagy levels in the LV tissues. TRF can attenuate cardiac remodeling in SHRs by regulating autophagy levels.
Collapse
Affiliation(s)
- Xin Yi
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department 1 of Cardiovasology, North China University of Science and Technology Affiliated Hospital, Tangshan City, 063000, Hebei Province, China
| | - Razif Abas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | | | - Jie Yan
- Department 1 of Cardiovasology, North China University of Science and Technology Affiliated Hospital, Tangshan City, 063000, Hebei Province, China
| | - Canzhang Liu
- Department 1 of Cardiovasology, North China University of Science and Technology Affiliated Hospital, Tangshan City, 063000, Hebei Province, China
| | - Jiaxu An
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ummi Nadira Daut
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
39
|
Liu C, Wang X, Xu S, Liu M, Cao X. Regulation of autophagy: Insights into O-GlcNAc modification mechanisms. Life Sci 2025; 369:123547. [PMID: 40058573 DOI: 10.1016/j.lfs.2025.123547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/30/2025]
Abstract
Autophagy is a "self-eating" biological process that degrades cytoplasmic contents to ensure cellular homeostasis. Its response to stimuli occurs in two stages: Within a few to several hours of exposure to a stress condition, autophagic flow rapidly increases, which is mediated by post-translational modification (PTM). Subsequently, the transcriptional program is activated and mediates the persistent autophagic response. O-linked β-N-acetylglucosamine (O-GlcNAc) modification is an inducible and dynamically cycling PTM; mounting evidence suggests that O-GlcNAc modification participates in the total autophagic process, including autophagy initiation, autophagosome formation, autophagosome-lysosome fusion, and transcriptional process. In this review, we summarize the current knowledge on the emerging role of O-GlcNAc modification in regulating autophagy-associated proteins and explain the different regulatory effects on autophagy exerted by O-GlcNAc modification.
Collapse
Affiliation(s)
- Chengzhi Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinyu Wang
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Shengnan Xu
- College of Basic Medicine, Dalian Medical University, Dalian 116044, China
| | - Mingyue Liu
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xusheng Cao
- Beijing Ophthalmology & Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China.
| |
Collapse
|
40
|
Acharya D, Sayyad Z, Hoenigsperger H, Hirschenberger M, Zurenski M, Balakrishnan K, Zhu J, Gableske S, Kato J, Zhang SY, Casanova JL, Moss J, Sparrer KMJ, Gack MU. TRIM23 mediates cGAS-induced autophagy in anti-HSV defense. Nat Commun 2025; 16:4418. [PMID: 40360474 PMCID: PMC12075517 DOI: 10.1038/s41467-025-59338-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
The cGAS-STING pathway, well-known to elicit interferon (IFN) responses, is also a key inducer of autophagy upon virus infection or other stimuli. Whereas the mediators for cGAS-induced IFN responses are well characterized, much less is known about how cGAS elicits autophagy. Here, we report that TRIM23, a unique TRIM protein harboring both ubiquitin E3 ligase and GTPase activity, is crucial for cGAS-STING-dependent antiviral autophagy. Genetic ablation of TRIM23 impairs autophagic control of HSV-1 infection. HSV-1 infection or cGAS-STING stimulation induces TBK1-mediated TRIM23 phosphorylation at S39, which triggers TRIM23 autoubiquitination and GTPase activity and ultimately elicits autophagy. Fibroblasts from a patient with herpes simplex encephalitis heterozygous for a dominant-negative, kinase-inactivating TBK1 mutation fail to activate autophagy by TRIM23 and cGAS-STING. Our results thus identify the cGAS-STING-TBK1-TRIM23 axis as a key autophagy defense pathway and may stimulate new therapeutic interventions for viral or inflammatory diseases.
Collapse
Affiliation(s)
- Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Zuberwasim Sayyad
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | | | | | - Matthew Zurenski
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Kannan Balakrishnan
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Junji Zhu
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Sebastian Gableske
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
- Eisai GmbH, Frankfurt am Main, Germany
| | - Jiro Kato
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Joel Moss
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Michaela U Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA.
- Department of Microbiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
41
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
42
|
Wu M, Yu C, Wen F, Li Y, Zhang X, Wang Y, Chen X, Chen X. NLRP3 inflammasome inhibits mitophagy during the progression of temporal lobe epilepsy. Sci Rep 2025; 15:16341. [PMID: 40348802 PMCID: PMC12065917 DOI: 10.1038/s41598-025-01087-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
Epilepsy is a neurological disorder involving mitochondrial dysfunction and neuroinflammation. This study examines the relationship between NLRP3 inflammasome activation and mitophagy in the temporal lobe epilepsy, which has not been reported before. A pilocarpine-induced epileptic rat model was used to assess seizure activity and neuronal loss. Pyroptosis markers (NLRP3, cleaved Gasdermin D, IL-1β/IL-18), and autophagy/mitophagy activity (LC3B-II/I, BNIP3, TOMM20/LC3B colocalization) were analyzed via immunofluorescence, Western blot, and transmission electron microscopy. NLRP3 inhibitors and anti-IL-1β antibodies were administered to evaluate therapeutic effects. Epileptic rats exhibited progressive neuronal loss and seizure aggravation, correlating with NLRP3 inflammasome activation and pyroptosis. While general autophagy was upregulated, mitophagy was selectively impaired in the hippocampus. NLRP3 activation promoted IL-1β release, which suppressed mitophagy via PPTC7 upregulation. NLRP3 activation inhibitor (MCC950) and anti-IL-1β treatment restored mitophagy and reduced seizures. NLRP3 inflammasome-driven pyroptosis exacerbates epilepsy by impairing mitophagy activity via IL-1β/PPTC7. Targeted NLRP3 inhibition mitigates this cascade, offering a promising strategy for refractory epilepsy.
Collapse
Affiliation(s)
- Mengqian Wu
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Cong Yu
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Fuli Wen
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Yunfei Li
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Xu Zhang
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Yinzhou Wang
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Xiaoqian Chen
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| | - Xingyong Chen
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
43
|
Li Y, Guo T, He J, Liu D, Peng S, Xu A. SLC35A2-mediated bisected GlcNAc-modified extracellular vesicles enhance immune regulation in breast cancer lung metastasis. Int Immunopharmacol 2025; 154:114505. [PMID: 40157085 DOI: 10.1016/j.intimp.2025.114505] [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: 12/07/2024] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
This study investigates the role of SLC35A2-mediated bisected GlcNAc-modified small extracellular vesicles (sEVs) in breast cancer (BC) lung metastasis. By modulating B3GALT1 expression, these sEVs regulate the pre-metastatic immune microenvironment, enhancing CD8+ T cell infiltration and reducing immune evasion. The use of β-peptide-loaded sEVs further amplifies anti-metastatic effects, as demonstrated in vivo mouse models and molecular analyses. These findings underscore the therapeutic potential of glycosylation-modified sEVs in enhancing immune responses and controlling BC metastasis.
Collapse
Affiliation(s)
- Yangyang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Tao Guo
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Juntong He
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Defeng Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Shihao Peng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
44
|
Guan D, Chen Z, Zhang Y, Sun W, Li L, Huang X. Dual Role of Natural Killer Cells in Early Pregnancy: Immunopathological Implications and Therapeutic Potential in Recurrent Spontaneous Abortion and Recurrent Implantation Failure. Cell Prolif 2025:e70037. [PMID: 40325291 DOI: 10.1111/cpr.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 05/07/2025] Open
Abstract
Natural killer (NK) cells are critical regulators of immune processes during early pregnancy, playing a key role in maintaining maternal-foetal immune tolerance and supporting successful implantation. In particular, uterine NK cells, a specialised subset of NK cells, facilitate trophoblast invasion, spiral artery remodelling and placental establishment. Dysregulation of NK cell activity, however, has been implicated in pregnancy complications, notably recurrent spontaneous abortion (RSA) and recurrent implantation failure (RIF). Aberrant NK cell functions, such as heightened cytotoxicity or defective immune signalling, can disrupt the balance between immune tolerance and response, leading to impaired placental development, reduced trophoblast activity and compromised uteroplacental blood flow. This review examines the role of NK cells in early pregnancy, emphasising their contributions to immune modulation and placentation. It also investigates the mechanisms by which NK cell dysfunction contributes to RSA and RIF, and explores therapeutic strategies aimed at restoring NK cell balance to improve pregnancy outcomes. A deeper understanding of NK cell interactions during early pregnancy may provide critical insights into the pathogenesis of pregnancy failure and facilitate targeted immunotherapeutic approaches.
Collapse
Affiliation(s)
- Defeng Guan
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhou Chen
- Gansu Provincial Hospital, Lanzhou, Gansu, China
- The Third Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yuhua Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wenjie Sun
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lifei Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xia Huang
- Gansu Provincial Hospital, Lanzhou, Gansu, China
- The Third Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
45
|
Lipovšek S, Vajs T, Dariš B, Novak T, Kozel P. Autophagic activity in the midgut cells of three arachnids responds selectively to different modes of overwintering in caves. PROTOPLASMA 2025; 262:531-544. [PMID: 39630263 DOI: 10.1007/s00709-024-02009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 11/14/2024] [Indexed: 04/24/2025]
Abstract
Autophagy is a highly conserved metabolic process that regulates cellular homeostasis and energy supply by degrading dysfunctional and excess cell constituents and reserve materials into products that are reused in metabolic and biosynthetic pathways. Macroautophagy is the best studied form of autophagy in invertebrates. Starvation is a common stress factor triggering autophagy in overwintering animals. In arachnids, the midgut diverticula cells perform many vital metabolic functions and are therefore critically involved in the response to starvation. Here we studied macroautophagy in three species which apply different modes for overwintering in caves: the harvestmen Gyas annulatus in diapause, Amilenus aurantiacus with ongoing ontogenesis under fasting conditions, and the spider Meta menardi, which feeds opportunistically even in winter. The main goal was to find eventual qualitative and quantitative differences in autophagic processes by inspecting TEM micrographs. In all three species, the rates of midgut epithelial cells with autophagic structures gradually increased during overwintering, but were significantly lower in G. annulatus in the middle and at the end of overwintering than in the other two species, owing to metabolic activity having been more suppressed. Decomposition of mitochondria and glycogen took place in autophagic structures in all three species. Moreover, spherite disintegration in A. aurantiacus and a special form of lipid disintegration through "lipid bubbly structures" in M. menardi indicate the crucial involvment of selective autophagy, while no specific autophagy was observed in G. annulatus. We conclude that autophagic activities support overwintering in different ways in the species studied.
Collapse
Affiliation(s)
- Saška Lipovšek
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000, Maribor, Slovenia
- Division of Cell Biology, Gottfried Schatz Research Center, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000, Maribor, Slovenia
| | - Tanja Vajs
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Barbara Dariš
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Tone Novak
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000, Maribor, Slovenia
| | - Peter Kozel
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000, Maribor, Slovenia.
- Research Centre of the Slovenian Academy of Science and Arts, Karst Research Institute, Titov trg 2, 6230, Postojna, Slovenia.
| |
Collapse
|
46
|
Bensalem J, Teong XT, Hattersley KJ, Hein LK, Fourrier C, Dang LVP, Singh S, Liu K, Wittert GA, Hutchison AT, Heilbronn LK, Sargeant TJ. Intermittent time-restricted eating may increase autophagic flux in humans: an exploratory analysis. J Physiol 2025; 603:3019-3032. [PMID: 40345145 DOI: 10.1113/jp287938] [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: 10/22/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Autophagy slows age-related pathologies and is stimulated by nutrient restriction in animal studies. However, this has never been shown in humans. We measured autophagy using a physiologically relevant measure of autophagic flux (flux of MAP1LC3B isoform II/LC3B-II in peripheral blood mononuclear cells in the context of whole blood) in 121 humans with obesity who were randomised to standard care (SC, control condition), calorie restriction (CR) or intermittent fasting plus time-restricted eating (iTRE) for 6 months. While the differences in change from baseline between groups was not significant at 2 months, we observed a significant difference in change from baseline between iTRE compared to SC at 6 months (P = 0.04, post hoc analysis). This effect may be driven partly by a tendency for autophagy to decrease in the SC group. The difference in change from baseline between CR and SC was not significant. Uncorrected analysis of correlations showed a negative relationship between change in autophagy and change in blood triglycerides. Data on the specificity and performance of the methods used to measure human autophagy are also presented. This shows autophagy may be increased by intermittent nutrient restriction in humans. If so, this is a demonstration that nutrient restriction can be used to improve a primary hallmark of biological ageing and provides a mechanism for how fasting could delay the onset of age-related disease. KEY POINTS: Autophagy slows biological ageing, and dysfunction of autophagy has been implicated in age-related disease - an effective way of increasing autophagy in cells and animal models is nutrient restriction. However, the impact of different types of nutrient restriction on physiological autophagic flux in humans has not been extensively researched. Here we measure the effect of intermittent time-restricted eating (iTRE) and calorie restriction on physiological autophagic flux in peripheral blood mononuclear cells. After 6 months, there was a significant difference in change from baseline between the iTRE and the standard care control group, with flux being higher in the iTRE group at this timepoint. However, there was no significant increase from baseline within the iTRE group, showing that although autophagy may be modified by nutrient restriction in humans, further studies are required.
Collapse
Affiliation(s)
- Julien Bensalem
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Xiao Tong Teong
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Kathryn J Hattersley
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Leanne K Hein
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Célia Fourrier
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Linh V P Dang
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Sanjna Singh
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | - Kai Liu
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Gary A Wittert
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Amy T Hutchison
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Leonie K Heilbronn
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Timothy J Sargeant
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| |
Collapse
|
47
|
Sisto A, van Wermeskerken T, Pancher M, Gatto P, Asselbergh B, Assunção Carreira ÁS, De Winter V, Adami V, Provenzani A, Timmerman V. Autophagy induction by piplartine ameliorates axonal degeneration caused by mutant HSPB1 and HSPB8 in Charcot-Marie-Tooth type 2 neuropathies. Autophagy 2025; 21:1116-1143. [PMID: 39698979 PMCID: PMC12013449 DOI: 10.1080/15548627.2024.2439649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
HSPB1 [heat shock protein family B (small) member 1] and HSPB8 are essential molecular chaperones for neuronal proteostasis, as they prevent protein aggregation. Mutant HSPB1 and HSPB8 primarily harm peripheral neurons, resulting in axonal Charcot-Marie-Tooth neuropathies (CMT2). Macroautophagy/autophagy is a shared mechanism by which HSPB1 and HSPB8 mutations cause neuronal dysfunction. Autophagosome formation is reduced in mutant HSPB1-induced pluripotent stem-cell-derived motor neurons from CMT type 2F patients. Likewise, the HSPB8K141N knockin mouse model, mimicking CMT type 2 L, exhibits axonal degeneration and muscle atrophy, with SQSTM1/p62-positive deposits. We show here that mouse embryonic fibroblasts isolated from a HSPB8K141N/green fluorescent protein (GFP)-LC3 model have diminished autophagosome production under conditions of MTOR inhibition. To correct the autophagic deficits in the HSPB1 and HSPB8 models, we screened by high-throughput autophagosome quantification the repurposing Spectrum Collection library for molecules that could boost the autophagic activity above the canonical MTOR inhibition. Hit compounds were validated on motor neurons obtained by differentiation of HSPB1P182L and HSPB8K141N patient-derived induced pluripotent stem cells, focusing on autophagy induction as well as neurite network density, axonal degeneration, and mitochondrial morphology. We identified molecules that specifically stimulate autophagosome formation in the HSPB8K141N cells, without affecting autophagy flux. Two top lead compounds induced autophagy and reduced axonal degeneration, thus promoting neuronal network maturation in the CMT2 patient-derived motor neurons. Based on these findings, the phenotypical screen revealed that piplartine rescued autophagy deficiencies in both the HSPB1 and HSPB8 models, demonstrating autophagy induction as an effective therapeutic strategy for CMT neuropathies and other chaperonopathies.
Collapse
Affiliation(s)
- Angela Sisto
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Tamira van Wermeskerken
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Pamela Gatto
- HTS Core Facility, University of Trento, Trento, Italy
| | - Bob Asselbergh
- Neuromics Support Facility, VIB - Center for Molecular Neurology, Antwerp, Belgium
- Neuromics Support Facility, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Vicky De Winter
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | | | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
48
|
Nàger M, Larsen KB, Bhujabal Z, Kalstad TB, Rössinger J, Myrmel T, Weinberger F, Birgisdottir AB. Mitophagy is induced in human engineered heart tissue after simulated ischemia and reperfusion. J Cell Sci 2025; 138:jcs263408. [PMID: 39912384 PMCID: PMC11959618 DOI: 10.1242/jcs.263408] [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] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
The paradoxical exacerbation of cellular injury and death during reperfusion remains a problem in the treatment of myocardial infarction. Mitochondrial dysfunction plays a key role in the pathogenesis of myocardial ischemia and reperfusion injury. Dysfunctional mitochondria can be removed by mitophagy, culminating in their degradation within acidic lysosomes. Mitophagy is pivotal in maintaining cardiac homeostasis and emerges as a potential therapeutic target. Here, we employed beating human engineered heart tissue (EHT) to assess mitochondrial dysfunction and mitophagy during ischemia and reperfusion simulation. Our data indicate adverse ultrastructural changes in mitochondrial morphology and impairment of mitochondrial respiration. Furthermore, our pH-sensitive mitophagy reporter EHTs, generated by a CRISPR/Cas9 endogenous knock-in strategy, revealed induced mitophagy flux in EHTs after ischemia and reperfusion simulation. The induced flux required the activity of the protein kinase ULK1, a member of the core autophagy machinery. Our results demonstrate the applicability of the reporter EHTs for mitophagy assessment in a clinically relevant setting. Deciphering mitophagy in the human heart will facilitate development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mireia Nàger
- Division of Cardiothoracic and Respiratory Medicine, University Hospital of North Norway, 9019 Tromsø, Norway
| | - Kenneth B. Larsen
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
- Department of Medical Biology, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Zambarlal Bhujabal
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Trine B. Kalstad
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Judith Rössinger
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Truls Myrmel
- Division of Cardiothoracic and Respiratory Medicine, University Hospital of North Norway, 9019 Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, 20251 Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Asa B. Birgisdottir
- Division of Cardiothoracic and Respiratory Medicine, University Hospital of North Norway, 9019 Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| |
Collapse
|
49
|
Apard T, Martinel V. Percutaneous ultrasound guided retrograde lacertus fibrosus release. HAND SURGERY & REHABILITATION 2025; 44S:102130. [PMID: 40139442 DOI: 10.1016/j.hansur.2025.102130] [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: 12/21/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 03/29/2025]
Abstract
The purpose of this technical note is to present a microinvasive percutaneous ultrasound-guided release of the lacertus fibrosus of the biceps brachii for lacertus syndrome, i.e., median nerve entrapment at the elbow. Using the Walant technique (wide awake local anesthesia and no tourniquet), the entry point is made with a 19G needle to introduce the hook distally from the distal border of the lacertus fibrosus. The hook is then slid along the pronator teres fascia to the proximal border of the lacertus fibrosus. The cut is ultrasound-guided from proximal to distal. The cut is effective when the back of the force is observed in the 3 targeted muscles (flexor carpi radialis, flexor pollicis longus and flexor digitorum profundus). This minimally invasive surgical procedure is efficient in terms of lacertus fibrosus sectioning. Real-time ultrasound monitoring may improve safety. The technique could be considered as a new ultrasound-guided alternative to open surgery. When performed superficially to the pronator teres muscle under WALANT anesthesia and percutaneously, ultrasound-guided lacertus fibrosus release may be an effective treatment for lacertus syndrome in the interventional ultrasound unit.
Collapse
Affiliation(s)
- Thomas Apard
- Ultrasound Guided Hand Surgery Center - Clinique Les Franciscaines of Versailles, 2 rue de Tocqueville, 78000 Versailles, France
| | - Vincent Martinel
- Orthopedic Group Ormeau Pyrénées, Polyclinique de l'Ormeau, ELSAN Tarbes, France
| |
Collapse
|
50
|
Yan Z, Han J, Mi Z, Wang Z, Fu Y, Wang C, Dang N, Liu H, Zhang F. GPNMB disrupts SNARE complex assembly to maintain bacterial proliferation within macrophages. Cell Mol Immunol 2025; 22:512-526. [PMID: 40038549 PMCID: PMC12041529 DOI: 10.1038/s41423-025-01272-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Xenophagy plays a crucial role in restraining the growth of intracellular bacteria in macrophages. However, the machinery governing autophagosome‒lysosome fusion during bacterial infection remains incompletely understood. Here, we utilize leprosy, an ideal model for exploring the interactions between host defense mechanisms and bacterial infection. We highlight the glycoprotein nonmetastatic melanoma protein B (GPNMB), which is highly expressed in macrophages from lepromatous leprosy (L-Lep) patients and interferes with xenophagy during bacterial infection. Upon infection, GPNMB interacts with autophagosomal-localized STX17, leading to a reduced N-glycosylation level at N296 of GPNMB. This modification promotes the degradation of SNAP29, thus preventing the assembly of the STX17-SNAP29-VAMP8 SNARE complex. Consequently, the fusion of autophagosomes with lysosomes is disrupted, resulting in inhibited cellular autophagic flux. In addition to Mycobacterium leprae, GPNMB deficiency impairs the proliferation of various intracellular bacteria in human macrophages, suggesting a universal role of GPNMB in intracellular bacterial infection. Furthermore, compared with their counterparts, Gpnmbfl/fl Lyz2-Cre mice presented decreased Mycobacterium marinum amplification. Overall, our study reveals a previously unrecognized role of GPNMB in host antibacterial defense and provides insights into its regulatory mechanism in SNARE complex assembly.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Jinghong Han
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zihao Mi
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenzhen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yixuan Fu
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Chuan Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ningning Dang
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Furen Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|