1
|
Diokmetzidou A, Scorrano L. Mitochondria-membranous organelle contacts at a glance. J Cell Sci 2025; 138:jcs263895. [PMID: 40357586 DOI: 10.1242/jcs.263895] [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] [Indexed: 05/15/2025] Open
Abstract
Mitochondrial contact sites are specialized interfaces where mitochondria physically interact with other organelles. Stabilized by molecular tethers and defined by unique proteomic and lipidomic profiles, these sites enable direct interorganellar communication and functional coordination, playing crucial roles in cellular physiology and homeostasis. Recent advances have expanded our knowledge of contact site-resident proteins, illuminated the dynamic and adaptive nature of these interfaces, and clarified their contribution to pathophysiology. In this Cell Science at a Glance article and the accompanying poster, we summarize the mitochondrial contacts that have been characterized in mammals, the molecular mechanisms underlying their formation, and their principal functions.
Collapse
Affiliation(s)
- Antigoni Diokmetzidou
- Department of Biology, University of Padova, 35121 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, 35121 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| |
Collapse
|
2
|
Zhuang J, Wang Y, Wu X, Peng Z, Huang Z, Zhao C, Shen B. SIGMAR1 screened by a GPCR-related classifier regulates endoplasmic reticulum stress in bladder cancer. J Transl Med 2025; 23:417. [PMID: 40211230 PMCID: PMC11987370 DOI: 10.1186/s12967-025-06393-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/17/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Bladder cancer (BC) is one of the most common malignancies worldwide. G protein-coupled receptors (GPCRs) are a large family of transmembrane proteins that are increasingly recognised as key players in cancer biology, affecting cell signalling and the tumour microenvironment. The sigma-1 receptor (SIGMAR1), although not a classical GPCR, has similar functions and is associated with the regulation of ER stress. However, its specific role and mechanism in bladder cancer are still unclear. METHOD The data sets pertaining to batch sequencing, single-cell RNA sequencing (scRNA-seq), immunotherapy response and clinical pathological characteristics were obtained from the public database. Thereafter, multiple algorithms were employed for the screening of GPCRs and immune cells related to the prognosis of BC. A GPCR-tumour microenvironment (TME) classifier was constructed and validated using different queues and multi-omics methods. The key biological pathways between GPCR-TME subgroups were identified through the utilisation of methodologies such as Gene Set Enrichment Analysis (GSEA), Weighted Gene Co-expression Network Analysis (WGCNA), and Tumour Immunophenotype Tracking (TIP). The expression of SIGMAR1 in BC cell lines and tissue samples was validated by western blotting. The Gene Ontology (GO) and GSEA were employed for biological process enrichment analysis. The biological role of SIGMAR1 in BC was investigated through functional experiments and subcutaneous tumour-bearing experiments in nude mice. The relationship between SIGMAR1 and immune cell infiltration was explored using the CIBERSORT method. RESULTS A total of 15 types of GPCR and 5 types of immune cells were identified and established as a GPCR-TME classifier. Patients in the GPCR-low + TME-high group exhibited the most favourable prognosis, whereas patients in the GPCR-high + TME-low group demonstrated the least favourable prognosis. The scRNA-seq results revealed an increase in GPCR expression in CD8 + T cells, endothelial cells, and NK cells. GPCR-TME was significantly correlated with overall survival (OS) in BC patients and outperformed a range of clinical parameters, making it an independent risk factor affecting the prognosis of BC patients. In comparison to normal tissues, SIGMAR1 was markedly expressed in BC tissues, and was associated with a poor prognosis. Functional experiments demonstrated that SIGMAR1 deficiency impeded the invasive capacity of cancer cells and restrained cellular proliferation. Moreover, in vivo experiments corroborated that SIGMAR1 deficiency curtailed the growth of xenografts in nude mice. Western blotting analysis revealed that SIGMAR1 silencing intensified endoplasmic reticulum (ER) stress in BC cells and promoted cell apoptosis. Additionally, the expression level of SIGMAR1 was correlated with the level of immune cell infiltration and immune-related functions. CONCLUSION The construction of a BC-related GPCR-TME classifier enabled the effective prediction of the OS of BC patients and the identification of SIGMAR1, a key factor regulating ER stress in BC. The knockout of SIGMAR1 can destroy its protective effect on ER stress, enhance apoptosis of BC cells, and facilitate further investigation of novel treatment strategies for cancer therapy.
Collapse
Affiliation(s)
- Jingming Zhuang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyong Wu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zijing Peng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengnan Huang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Zhao
- MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences & National Clinical Research Center for Aging and Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China.
| | - Bing Shen
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Urology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, China.
| |
Collapse
|
3
|
Salvador-Mira M, Gimenez-Moya P, Manso-Aznar A, Sánchez-Córdoba E, Sevilla-Diez MA, Chico V, Nombela I, Puente-Marin S, Roher N, Perez L, Dučić T, Benseny-Cases N, Perez-Berna AJ, Ortega-Villaizan MDM. Viral vaccines promote endoplasmic reticulum stress-induced unfolding protein response in teleost erythrocytes. Eur J Cell Biol 2025; 104:151490. [PMID: 40252498 DOI: 10.1016/j.ejcb.2025.151490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025] Open
Abstract
Most available evidence points to a proviral role for endoplasmic reticulum (ER) stress, as many viruses exploit it to promote viral replication. In contrast, few studies have linked ER stress to the antiviral immune response, and even fewer to the vaccine-induced immune response. In this work, we demonstrated that ER stress is a key molecular link in the immune response of teleost erythrocytes or red blood cells (RBCs) under vaccine stimulation. Moreover, the unfolded protein response (UPRER) triggered by ER stress may work together with autophagy and related cellular mechanisms as part of a coordinated immune response in RBCs. We unveiled biochemical changes in the lipid-protein profile of vaccine-treated RBCs by synchrotron radiation-based Fourier transform infrared microspectroscopy (SR-µFTIR) associated with the modulation of ER expansion, increased mitochondrial number, and vesicular structures detected by soft X-ray cryotomography (cryo-SXT). We found a positive correlation between both morphological and biochemical changes and the expression of genes related to UPRER, autophagy, mitochondrial stress, vesicle trafficking, and extracellular vesicle release. These processes in RBCs are ideal cellular targets for the development of more specific prophylactic tools with greater immunogenic capacity than currently available options.
Collapse
Affiliation(s)
- Maria Salvador-Mira
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Paula Gimenez-Moya
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Alba Manso-Aznar
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Ester Sánchez-Córdoba
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Manuel A Sevilla-Diez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Veronica Chico
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Ivan Nombela
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Sara Puente-Marin
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Nerea Roher
- Institute of Biotechnology and Biomedicine (IBB) & Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luis Perez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain
| | - Tanja Dučić
- ALBA Synchrotron Light Source, Cerdanyola del Vallès, Barcelona, Spain
| | - Núria Benseny-Cases
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Maria Del Mar Ortega-Villaizan
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (IDiBE-UMH), Elche, Spain.
| |
Collapse
|
4
|
Kong X, Liu T, Wei J. Parkinson's Disease: The Neurodegenerative Enigma Under the "Undercurrent" of Endoplasmic Reticulum Stress. Int J Mol Sci 2025; 26:3367. [PMID: 40244210 PMCID: PMC11989508 DOI: 10.3390/ijms26073367] [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: 03/11/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Parkinson's disease (PD), a prevalent neurodegenerative disorder, demonstrates the critical involvement of endoplasmic reticulum stress (ERS) in its pathogenesis. This review comprehensively examines the role and molecular mechanisms of ERS in PD. ERS represents a cellular stress response triggered by imbalances in endoplasmic reticulum (ER) homeostasis, induced by factors such as hypoxia and misfolded protein aggregation, which activate the unfolded protein response (UPR) through the inositol-requiring enzyme 1 (IRE1), protein kinase R-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6) pathways. Clinical, animal model, and cellular studies have consistently demonstrated a strong association between PD and ERS. Abnormal expression of ERS-related molecules in PD patients' brains and cerebrospinal fluid (CSF) correlates with disease progression. In animal models (e.g., Drosophila and mice), ERS inhibition alleviates dopaminergic neuronal damage. Cellular experiments reveal that PD-mimicking pathological conditions induce ERS, while interactions between ERS and mitochondrial dysfunction promote neuronal apoptosis. Mechanistically, (1) pathological aggregation of α-synuclein (α-syn) and ERS mutually reinforce dopaminergic neuron damage; (2) leucine-rich repeat kinase 2 (LRRK2) gene mutations induce ERS through thrombospondin-1 (THBS1)/transforming growth factor beta 1 (TGF-β1) interactions; (3) molecules such as Parkin and PTEN-induced kinase 1 (PINK1) regulate ERS in PD. Furthermore, ERS interacts with mitochondrial dysfunction, oxidative stress, and neuroinflammation to exacerbate neuronal injury. Emerging therapeutic strategies show significant potential, including artificial intelligence (AI)-assisted drug design targeting ERS pathways and precision medicine approaches exploring non-pharmacological interventions such as personalized electroacupuncture. Future research should focus on elucidating ERS-related mechanisms and identifying novel therapeutic targets to develop more effective treatments for PD patients, ultimately improving their quality of life.
Collapse
Affiliation(s)
- Xiangrui Kong
- Wushu College, Henan University, Kaifeng 475004, China;
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China;
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China;
| | - Jianshe Wei
- Wushu College, Henan University, Kaifeng 475004, China;
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China;
| |
Collapse
|
5
|
Liu Z, Ha DP, Lin LL, Qi L, Lee AS. Requirements for nuclear GRP78 transcriptional regulatory activities and interaction with nuclear GRP94. J Biol Chem 2025; 301:108369. [PMID: 40024475 PMCID: PMC11997380 DOI: 10.1016/j.jbc.2025.108369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
GRP78, a molecular chaperone primarily located in the endoplasmic reticulum (ER), has recently been discovered to translocate into the nucleus of stressed and cancer cells where it assumes a new function reprogramming the transcriptome. This study explores the requirements of GRP78 nuclear translocation and its transcriptional activity and investigates the role of ER-associated degradation in the process. We show that the ER-processed, mature form of GRP78 is the major form of nuclear GRP78 and is the form with transcriptional regulatory activity. In contrast, exogenously expressed GRP78 designed to lack its ER signal peptide, thus preventing it from entering the ER or undergoing any ER-related processing/modification, while able to enter the nucleus, lacks transcriptional regulatory activity toward E-Box containing target genes. Additionally, the ATP-binding and substrate-binding activities of GRP78 are critical for this transcriptional regulatory function. We further discover that GRP94, an ER chaperone that acts in concert with GRP78 on protein folding, can translocate to the nucleus and colocalize with nuclear GRP78 upon ER stress. These findings suggest that some form of ER processing of GRP78, in addition to cleavage of the ER signal peptide, is critical for its nuclear activity and that in stressed cells, ER chaperones may assume new functions in the nucleus yet to be explored.
Collapse
Affiliation(s)
- Ze Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, Virginia, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, Virginia, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
6
|
Liu Y, Qiu S, Huang H, Wu Z, Ge S. Ferrostatin supplementation improves microalgal activities and nutrient removal in wastewater under high temperature shock: From ferroptosis-like inhibition to enhanced oxidation resistance. WATER RESEARCH 2025; 273:123033. [PMID: 39721506 DOI: 10.1016/j.watres.2024.123033] [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: 10/18/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
High temperature (HT) shock is one of environmental stressors suppressing microalgal activities in microalgal wastewater bioremediation system. However, its inhibition mechanism and how to alleviate such suppression remain inadequately understood. This study confirmed a transient ferroptosis as a novel form of programmed cell death in a wastewater-indigenous Chlorella sp., responding to a 30-minute HT (50 °C) exposure, through the systematically physiological, metabolomic and transcriptomic analysis. Specifically, the HT-induced ferroptosis could be supported by both the growth and physiological indicators. These include the suppressed growth (76.05 %), suppressed nutrient removals (NH4+-N by 76.22 %, PO43--P by 64.15 %), accumulated intracellular Fe3+ concentrations (7.75-fold), enhanced oxidative stress (e.g., increased levels of reactive oxygen species (159.97 %)), activated antioxidant defense system (e.g., increased activities of superoxide dismutase (24.83 %) and catalase (5.03-fold)), and obvious membrane damage (e.g., increased levels of malondialdehyde (1.67-fold)). Further metabolomic analysis indicated that such HT-induced ferroptosis was also largely related to the significant alternations of lipid remodeling in three aspects: varied abundance of certain lipids specific to chloroplast membrane or mitochondria, accumulation of certain lipids with lower unsaturation, and formation of lipid peroxides disrupting membrane integrity. Moreover, the key genes involved in ferroptosis correspondingly responded, especially those associated with lipid metabolism (e.g., ACSL), antioxidant defense system (e.g., GSS, GPX and GSR), mitochondrial normal functioning (e.g., SEL1L), autophagy regulation (e.g., ATG9, ATG11, ATG13) and protein folding (e.g., HSPA5, HSPA1s, HSP90B). In addition, the supplementation of the typical ferroptosis inhibitor Ferrostatin-1 effectively mitigated lipid peroxide accumulation and suppressed the onset of ferroptosis, accelerating subsequent recovery of NH4+-N removal by 60.66 %. These findings update current understandings of microalgal ferroptosis-like inhibition, offering Ferrostatin-1 supplementation as a potential strategy for system resistance to heat stress in microalgae-based bioremediation system.
Collapse
Affiliation(s)
- Yanlin Liu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Shuang Qiu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Hongtao Huang
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Zhengshuai Wu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Shijian Ge
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China.
| |
Collapse
|
7
|
He P, Chang H, Qiu Y, Wang Z. Mitochondria associated membranes in dilated cardiomyopathy: connecting pathogenesis and cellular dysfunction. Front Cardiovasc Med 2025; 12:1571998. [PMID: 40166597 PMCID: PMC11955654 DOI: 10.3389/fcvm.2025.1571998] [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: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, yet therapeutic options remain limited. While traditional research has focused on mechanisms such as energy deficits and calcium dysregulation, increasing evidence suggests that mitochondria-associated membranes (MAMs) could provide new insights into understanding and treating DCM. In this narrative review, we summarize the key role of MAMs, crucial endoplasmic reticulum (ER)-mitochondria interfaces, in regulating cellular processes such as calcium homeostasis, lipid metabolism, and mitochondrial dynamics. Disruption of MAMs function may initiate pathological cascades, including ER stress, inflammation, and cell death. These disruptions in MAM function lead to further destabilization of cellular homeostasis. Identifying MAMs as key modulators of cardiac health may provide novel insights for early diagnosis and targeted therapies in DCM.
Collapse
Affiliation(s)
- Pingge He
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hongbo Chang
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yueqing Qiu
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhentao Wang
- Second School of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
8
|
Qiu J, Khedr MA, Pan M, Ferreira CR, Chen J, Snyder MM, Ajuwon KM, Yue F, Kuang S. Ablation of FAM210A in Brown Adipocytes of Mice Exacerbates High-Fat Diet-Induced Metabolic Dysfunction. Diabetes 2025; 74:282-294. [PMID: 39602358 PMCID: PMC11842609 DOI: 10.2337/db24-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Thermogenesis of brown adipose tissue (BAT) provides metabolic benefits against pathologic conditions, such as type 2 diabetes, obesity, cardiovascular disease, and cancer. The thermogenic function of BAT relies on mitochondria, but whether mitochondrial remodeling is required for the beneficial effects of BAT remains unclear. We recently identified FAM210A as a BAT-enriched mitochondrial protein essential for cold-induced thermogenesis through the modulation of OPA1-dependent cristae remodeling. Here, we report a key role of FAM210A in the systemic response to a high-fat diet (HFD). We discovered that an HFD suppressed FAM210A expression, associated with excessive OPA1 cleavage in BAT. Ucp1-Cre-driven BAT-specific Fam210a knockout (Fam210aUKO) similarly elevated OPA1 cleavage, accompanied by whitening of BAT. When subjected to an HFD, Fam210aUKO mice gained similar fat mass as sibling control mice but developed glucose intolerance, insulin resistance, and liver steatosis. The metabolic dysfunction was associated with overall increased lipid content in both the liver and BAT. Additionally, Fam210aUKO leads to inflammation in white adipose tissue. These data demonstrate that FAM210A in BAT is necessary for counteracting HFD-induced metabolic dysfunction but not obesity. ARTICLE HIGHLIGHTS FAM210A regulates cold-induced mitochondrial remodeling through control of OPA1 cleavage, but whether it also plays a role in high-fat diet (HFD)-induced cristae remodeling is unknown. We asked if an HFD would alter the FAM210A level and OPA1 cleavage in brown adipose tissue (BAT) and how FAM210A loss of function would affect diet-induced obesity in mice. We found that an HFD diminished FAM210A expression and accelerated OPA1 cleavage in BAT, and Fam210a knockout exacerbated HFD-induced whitening of BAT, cold intolerance, liver steatosis, white adipose tissue inflammation, and metabolic dysfunction. Our work reveals a physiologic role of FAM210A-mediated BAT mitochondrial remodeling in systemic adaptation to an HFD and suggests that BAT mitochondria may be targeted to treat diet-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Mennatallah A. Khedr
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Meijin Pan
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | | | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Madigan M. Snyder
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Biological Sciences, Purdue University, West Lafayette, IN
| | - Kolapo M. Ajuwon
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
- Institute for Cancer Research, Purdue University, West Lafayette, IN
| |
Collapse
|
9
|
Li Z, Ran Q, Qu C, Hu S, Cui S, Zhou Y, Shen B, Yang B. Sigma-1 receptor activation attenuates DOX-induced cardiotoxicity by alleviating endoplasmic reticulum stress and mitochondrial calcium overload via PERK and IP3R-VDAC1-MCU signaling pathways. Biol Direct 2025; 20:23. [PMID: 40001213 PMCID: PMC11853590 DOI: 10.1186/s13062-025-00617-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Doxorubicin (DOX) is an anthracycline with potent antitumor properties and rare yet serious cardiotoxic side effects that limit its clinical application. The sigma-1 receptor is a stress-triggered chaperone often dysregulated in diseases and has known cardioprotective effects. Although its anti-oxidative stress and anti-apoptotic effects have been demonstrated, its effectiveness in DOX-induced cardiotoxicity has never been explored. This study investigated the potential role of the activated sigma-1 receptor in a DOX-induced murine cardiotoxicity model to elucidate the receptor's mechanism of action. METHODS We established the model in C57BL/6 mice by daily intraperitoneal injections of fluvoxamine (Flv) for 4 consecutive weeks to activate the receptor and by weekly intraperitoneal injections of DOX at 5 mg/kg for 3 weeks. We performed in vitro experiments using cardiomyocytes of neonatal Sprague-Dawley rats to verify the protective effect of the sigma-1 receptor. RESULTS We found that sigma-1 expression in the heart decreased in the DOX-treated mice, and activating the receptor with Flv improved cardiac function. Moreover, Flv pretreatment inhibited cardiomyocyte apoptosis and endoplasmic reticulum stress and increased the expression of the Bcl2 apoptosis regulator (Bcl2), effectively alleviating the pathophysiological manifestations in mice. In addition, activating the receptor exerted cardioprotective effects by modulating endoplasmic reticulum stress through the PRKR-like endoplasmic reticulum kinase (PERK) signaling pathway. It also reduced mitochondrial and endoplasmic reticulum contact and alleviated mitochondrial calcium overload through the IP3R-VDAC1-MCU signaling pathway. CONCLUSION In conclusion, our study emphasizes the therapeutic potential of activating sigma-1 receptors against DOX-induced cardiotoxicity, suggesting sigma-1 receptors as potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Qian Ran
- Department of Cardiology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shengyu Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - You Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Bo Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| |
Collapse
|
10
|
Wu SA, Li ZJ, Qi L. Endoplasmic reticulum (ER) protein degradation by ER-associated degradation and ER-phagy. Trends Cell Biol 2025:S0962-8924(25)00002-9. [PMID: 39909774 DOI: 10.1016/j.tcb.2025.01.002] [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: 08/09/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 02/07/2025]
Abstract
Protein misfolding and aggregation in the endoplasmic reticulum (ER) have been causally linked to a variety of human diseases. Two key pathways for eliminating misfolded proteins and aggregates in the ER are ER-associated degradation (ERAD) and ER-phagy, respectively. While both pathways have been well characterized biochemically, our understanding of their physiological relevance and significance remains limited. In recent years, significant advances have been made, including the generation and characterization of various knockout and knockin mouse models, the identification of human disease-associated or -causing variants, and insights into the coordination between ERAD and autophagy in physiological contexts. In this review, we summarize these advancements, highlighting the key roles of a highly conserved suppressor of lin-12-like-hydroxymethyl glutaryl-coenzyme A reductase degradation 1 (SEL1L-HRD1) protein complex of ERAD and ER-phagy in health and disease.
Collapse
Affiliation(s)
- Shuangcheng Alivia Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA; Medical Scientist Training Program, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| |
Collapse
|
11
|
Zhao WB, Sheng R. The correlation between mitochondria-associated endoplasmic reticulum membranes (MAMs) and Ca 2+ transport in the pathogenesis of diseases. Acta Pharmacol Sin 2025; 46:271-291. [PMID: 39117969 PMCID: PMC11756407 DOI: 10.1038/s41401-024-01359-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Mitochondria and the endoplasmic reticulum (ER) are vital organelles that influence various cellular physiological and pathological processes. Recent evidence shows that about 5%-20% of the mitochondrial outer membrane is capable of forming a highly dynamic physical connection with the ER, maintained at a distance of 10-30 nm. These interconnections, known as MAMs, represent a relatively conserved structure in eukaryotic cells, acting as a critical platform for material exchange between mitochondria and the ER to maintain various aspects of cellular homeostasis. Particularly, ER-mediated Ca2+ release and recycling are intricately associated with the structure and functionality of MAMs. Thus, MAMs are integral in intracellular Ca2+ transport and the maintenance of Ca2+ homeostasis, playing an essential role in various cellular activities including metabolic regulation, signal transduction, autophagy, and apoptosis. The disruption of MAMs observed in certain pathologies such as cardiovascular and neurodegenerative diseases as well as cancers leads to a disturbance in Ca2+ homeostasis. This imbalance potentially aggravates pathological alterations and disease progression. Consequently, a thorough understanding of the link between MAM-mediated Ca2+ transport and these diseases could unveil new perspectives and therapeutic strategies. This review focuses on the changes in MAMs function during disease progression and their implications in relation to MAM-associated Ca2+ transport.
Collapse
Affiliation(s)
- Wen-Bin Zhao
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
12
|
Liu H, Cheng J, Ye F, Dong X, Ge W, Wang X, Zhao Y, Dan G, Chen M, Sai Y. Asiatic acid improves the damage of HaCaT cells induced by nitrogen mustard through inhibiting endoplasmic reticulum stress. Toxicol Res (Camb) 2025; 14:tfaf019. [PMID: 39968516 PMCID: PMC11831031 DOI: 10.1093/toxres/tfaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/09/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Nitrogen mustard (NM) belongs to vesicant agents. Blisters are one of the important characteristics of NM skin damage. It is urgent to further elucidate the mechanism and develop effective countermeasures for the skin damage induced by NM. The endoplasmic reticulum (ER) is an important intracellular organelle, playing an important role in maintaining cellular homeostasis. In this study, we explored the role of endoplasmic reticulum stress (ERS) and the protective effect of asiatic acid (AA) in the HaCaT cells induced by NM. It was found that the key regulatory proteins of ERS, such as glucose regulated protein 78 (GRP78), X-box binding protein 1 (XBP1), inositol requiring enzyme 1 (IRE1), Phospho-IRE1 (pIRE1), and TNF receptor associated factor 2 (TRAF2) were increased respectively in HaCaT cells exposed to NM compared with those of the control group, showing an increasing trend with the increase of NM exposure concentration and exposure time. Additionally, the protein expression of Caspase-3 and the Cleaved-Caspase-3 was also increased by NM in HaCaT cells, resulting in the apoptosis of HaCaT cells. Meanwhile, the content of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) was also increased in HaCaT cells exposed to NM. Further study showed that AA pretreatment could decrease the protein expression of GRP78, XBP1 and IRE1, pIRE1, TRAF2, Caspase-3, and Cleaved-Caspase-3. And moreover, AA also could reduce the content of TNF-α and IL-6. Overall, the present study showed that AA played an important protective effect in HaCaT cells exposed to NM through the inhibition of the ERS-induced apoptosis and inflammatory response.
Collapse
Affiliation(s)
- Haoyin Liu
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Jin Cheng
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Feng Ye
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Xunhu Dong
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Wei Ge
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Xiaogang Wang
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Yuanpeng Zhao
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Guorong Dan
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Mingliang Chen
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| | - Yan Sai
- Institute of Toxicology, Third Military Medical University, No. 30 Gaotanyan Avenue, Shapingba District, Chongqing 400038, China
| |
Collapse
|
13
|
Pang Y, Qin Y, Du Z, Liu Q, Zhang J, Han K, Lu J, Yuan Z, Li J, Pan S, Dong X, Xu M, Wang D, Li S, Li Z, Chen Y, Zhao Z, Zhang Z, Chuan S, Song Y, Sun M, Jia X, Xia Z, Zhan L, Yue Z, Cui W, Wang J, Gu Y, Ni M, Yang H, Xu X, Liu X, Li Q, Fan G. Single-cell transcriptome atlas of lamprey exploring Natterin- induced white adipose tissue browning. Nat Commun 2025; 16:752. [PMID: 39820434 PMCID: PMC11739602 DOI: 10.1038/s41467-025-56153-w] [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: 03/28/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Lampreys are early jawless vertebrates that are the key to understanding the evolution of vertebrates. However, the lack of cytomic studies on multiple lamprey organs has hindered progress in this field. Therefore, the present study constructed a comprehensive cell atlas comprising 604,460 cells/nuclei and 70 cell types from 14 lamprey tissue samples. Comparison of cellular evolution across species revealed that most lamprey cell types are homologous to those in jawed vertebrates. We discovered acinar- and islet-like cell populations despite the lack of parenchymal organs in lampreys, providing evidence of pancreatic function in vertebrates. Furthermore, we investigated the heterogeneity of lamprey immune cell populations. Natterin was highly expressed in granulocytes, and NATTERIN was localized to the lipid droplets. Moreover, we developed a transgenic mouse model expressing Natterin to elucidate the role of NATTERIN in lipid metabolism, whereas the browning of white adipose tissue was induced. These findings elucidate vertebrate cellular evolution and advance our understanding of adipose tissue plasticity and metabolic regulation in lampreys.
Collapse
Affiliation(s)
- Yue Pang
- College of Life Science, Liaoning Normal University, Dalian, 116081, China.
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Yating Qin
- BGI Research, Qingdao, 266555, China
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
- BGI Research, Hangzhou, 310030, China
| | - Zeyu Du
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Qun Liu
- BGI Research, Qingdao, 266555, China
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Jin Zhang
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Kai Han
- BGI Research, Qingdao, 266555, China
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Jiali Lu
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Zengbao Yuan
- BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Li
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | | | - Xinrui Dong
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Mengyang Xu
- BGI Research, Qingdao, 266555, China
- BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of marine biology genomics, BGI Research, Shenzhen, 518083, China
| | - Dantong Wang
- BGI Research, Qingdao, 266555, China
- BGI Research, Shenzhen, 518083, China
| | - Shuo Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, 266071, China
| | - Zhen Li
- BGI Research, Qingdao, 266555, China
| | | | - Zhisheng Zhao
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | | | - Shunqin Chuan
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Yue Song
- BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Mingjie Sun
- College of Life Science, Liaoning Normal University, Dalian, 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Zhangyong Xia
- Department of Neurology, The Second People's Hospital of Liaocheng, Liaocheng, Shandong, 252000, China
| | | | - Zhen Yue
- BGI Research, Sanya, 572025, China
| | - Wei Cui
- BGI Research, Qingdao, 266555, China
| | - Jun Wang
- BGI Research, Qingdao, 266555, China
| | - Ying Gu
- BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Ming Ni
- MGI Tech, Shenzhen, 518083, China
| | - Huanming Yang
- BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xin Liu
- BGI Research, Shenzhen, 518083, China.
- BGI, Shenzhen, 518083, China.
| | - Qingwei Li
- College of Life Science, Liaoning Normal University, Dalian, 116081, China.
- Lamprey Research Center, Liaoning Normal University, Dalian, 116081, China.
| | - Guangyi Fan
- BGI Research, Qingdao, 266555, China.
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- BGI Research, Hangzhou, 310030, China.
- BGI Research, Shenzhen, 518083, China.
- Shenzhen Key Laboratory of marine biology genomics, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
| |
Collapse
|
14
|
Norouzi Esfahani E, Knedlik T, Shin SH, Magalhães Rebelo AP, De Mario A, Vianello C, Persano L, Rampazzo E, Edomi P, Bean C, Brunetti D, Scorrano L, Greco S, Gerdol M, Giacomello M. Remodeling of Mitochondria-Endoplasmic Reticulum Contact Sites Accompanies LUHMES Differentiation. Biomolecules 2025; 15:126. [PMID: 39858520 PMCID: PMC11764118 DOI: 10.3390/biom15010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Neural progenitor cells (NPCs) are often used to study the subcellular mechanisms underlying differentiation into neurons in vitro. Works published to date have focused on the pathways that distinguish undifferentiated NPCs from mature neurons, neglecting the earlier and intermediate stages of this process. Current evidence suggests that mitochondria interaction with the ER is fundamental to a wide range of intracellular processes. However, it is not clear whether and how the mitochondria-ER interactions differ between NPCs and their differentiated counterparts. Here we take advantage of the widely used NPC line LUHMES to provide hints on the mitochondrial dynamic trait changes that occur during the first stage of their maturation into dopaminergic-like neurons. We observed that the morphology of mitochondria, their interaction with the ER, and the expression of several mitochondria-ER contact site resident proteins change, which suggests the potential contribution of mitochondria dynamics to NPC differentiation. Further studies will be needed to explore in depth these changes, and their functional outcomes, which may be relevant to the scientific community focusing on embryonic neurogenesis and developmental neurotoxicity.
Collapse
Affiliation(s)
- Emad Norouzi Esfahani
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
| | - Tomas Knedlik
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
| | - Sang Hun Shin
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
| | - Ana Paula Magalhães Rebelo
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
| | - Agnese De Mario
- Department of Biomedical Science, University of Padua, 35131 Padua, Italy;
| | - Caterina Vianello
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
| | - Luca Persano
- Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy; (L.P.); (E.R.)
- Pediatric Research Institute, Città della Speranza Foundation, 35127 Padua, Italy
| | - Elena Rampazzo
- Department of Women’s and Children’s Health, University of Padua, 35128 Padua, Italy; (L.P.); (E.R.)
- Pediatric Research Institute, Città della Speranza Foundation, 35127 Padua, Italy
| | - Paolo Edomi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (P.E.); (S.G.); (M.G.)
| | - Camilla Bean
- Department of Medicine, University of Udine, 33100 Udine, Italy;
| | - Dario Brunetti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico “C. Besta”, 20126 Milan, Italy;
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Luca Scorrano
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy
| | - Samuele Greco
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (P.E.); (S.G.); (M.G.)
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (P.E.); (S.G.); (M.G.)
| | - Marta Giacomello
- Department of Biology, University of Padua, 35131 Padua, Italy; (E.N.E.); (T.K.); (S.H.S.); (A.P.M.R.); (C.V.); (L.S.)
| |
Collapse
|
15
|
Li Y, Qi J, Guo L, Jiang X, He G. Organellar quality control crosstalk in aging-related disease: Innovation to pave the way. Aging Cell 2025; 24:e14447. [PMID: 39668579 PMCID: PMC11709098 DOI: 10.1111/acel.14447] [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: 09/15/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
Organellar homeostasis and crosstalks within a cell have emerged as essential regulatory and determining factors for the survival and functions of cells. In response to various stimuli, cells can activate the organellar quality control systems (QCS) to maintain homeostasis. Numerous studies have demonstrated that dysfunction of QCS can lead to various aging-related diseases such as neurodegenerative, pulmonary, cardiometabolic diseases and cancers. However, the interplay between QCS and their potential role in these diseases are poorly understood. In this review, we present an overview of the current findings of QCS and their crosstalk, encompassing mitochondria, endoplasmic reticulum, Golgi apparatus, ribosomes, peroxisomes, lipid droplets, and lysosomes as well as the aberrant interplays among these organelles that contributes to the onset and progression of aging-related disorders. Furthermore, potential therapeutic approaches based on these quality control interactions are discussed. Our perspectives can enhance insights into the regulatory networks underlying QCS and the pathology of aging and aging-related diseases, which may pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Yu Li
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Jinxin Qi
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
| | - Linhong Guo
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xian Jiang
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Gu He
- Department of Dermatology & VenerologyWest China Hospital, Sichuan UniversityChengduChina
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease‐Related Molecular Network, State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
16
|
Congur I, Mingrone G, Guan K. Targeting endoplasmic reticulum stress as a potential therapeutic strategy for diabetic cardiomyopathy. Metabolism 2025; 162:156062. [PMID: 39515414 DOI: 10.1016/j.metabol.2024.156062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/02/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Endoplasmic reticulum (ER) is an essential organelle involved in vesicular transport, calcium handling, protein synthesis and folding, and lipid biosynthesis and metabolism. ER stress occurs when ER homeostasis is disrupted by the accumulation of unfolded and/or misfolded proteins in the ER lumen. Adaptive pathways of the unfolded protein response (UPR) are activated to maintain ER homeostasis. In obesity and type 2 diabetes mellitus (T2DM), accumulating data indicate that persistent ER stress due to maladaptive UPR interacts with insulin/leptin signaling, which may be the potential and central mechanistic link between obesity-/T2DM-induced metabolic dysregulation (chronic hyperglycemia, dyslipidemia and lipotoxicity in cardiomyocytes), insulin/leptin resistance and the development of diabetic cardiomyopathy (DiabCM). Meanwhile, these pathological conditions further exacerbate ER stress. However, their interrelationships and the underlying molecular mechanisms are not fully understood. A deeper understanding of ER stress-mediated pathways in DiabCM is needed to develop novel therapeutic strategies. The aim of this review is to discuss the crosstalk between ER stress and leptin/insulin signaling and their involvement in the development of DiabCM focusing on mitochondria-associated ER membranes and chronic inflammation. We also present the current direction of drug development and important considerations for translational research into targeting ER stress for the treatment of DiabCM.
Collapse
Affiliation(s)
- Irem Congur
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany
| | - Geltrude Mingrone
- Division of Diabetes & Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, United Kingdom; Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany.
| |
Collapse
|
17
|
Kaniuka O, Deregowska A, Bandura Y, Sabadashka M, Chala D, Kulachkovskyi O, Kubis H, Adamczyk-Grochala J, Sybirna N. Upregulation of GRP78 is accompanied by decreased antioxidant response and mitophagy promotion in streptozotocin-induced type 1 diabetes in rats. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167531. [PMID: 39353543 DOI: 10.1016/j.bbadis.2024.167531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Endoplasmic reticulum stress, oxidative stress, and mitochondrial dysfunction are interconnected processes involved in the pathogenesis of diabetes mellitus (DM). In the present study, we demonstrate a distinct unfolded protein response (UPR) signaling pathways in two mammalian models of DM: β-TC-6 cell line and streptozotocin-induced type 1 diabetes model in rats. However, a feature common to both systems was the upregulation of the GRP78 protein. Moreover, in vivo studies showed the disruption of the antioxidant system and an escalation of mitophagy against the background of a depletion of the level of ATP in pancreatic cells. In conclusion, we suggest that glucotoxic conditions induced GRP78 upregulation, and next cause depletion of the antioxidant pool and disruption of the functioning of antioxidant defense enzymes and in consequence promote mitophagy in pancreatic cells. Therefore, GRP78 may be considered as a potential therapeutic factor in patients with diabetes.
Collapse
Affiliation(s)
- O Kaniuka
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - A Deregowska
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - Yu Bandura
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - M Sabadashka
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - D Chala
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - O Kulachkovskyi
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| | - H Kubis
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - J Adamczyk-Grochala
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - N Sybirna
- Department of Biochemistry, Ivan Franko National University of Lviv, 1, Universytetska St., 79000 Lviv, Ukraine.
| |
Collapse
|
18
|
Li H, Sun W, Gong W, Han Y. Transfer and fates of damaged mitochondria: role in health and disease. FEBS J 2024; 291:5342-5364. [PMID: 38545811 DOI: 10.1111/febs.17119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/27/2024] [Accepted: 03/04/2024] [Indexed: 12/19/2024]
Abstract
Intercellular communication is pivotal in mediating the transfer of mitochondria from donor to recipient cells. This process orchestrates various biological functions, including tissue repair, cell proliferation, differentiation and cancer invasion. Typically, dysfunctional and depolarized mitochondria are eliminated through intracellular or extracellular pathways. Nevertheless, increasing evidence suggests that intercellular transfer of damaged mitochondria is associated with the pathogenesis of diverse diseases. This review investigates the prevalent triggers of mitochondrial damage and the underlying mechanisms of mitochondrial transfer, and elucidates the role of directional mitochondrial transfer in both physiological and pathological contexts. Additionally, we propose potential previously unknown mechanisms mediating mitochondrial transfer and explore their prospective roles in disease prevention and therapy.
Collapse
Affiliation(s)
- Hanbing Li
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Weiyun Sun
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenwen Gong
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yubing Han
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Advanced Biomedical Imaging Facility-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Zhang R, Yang H, Guo M, Niu S, Xue Y. Mitophagy and its regulatory mechanisms in the biological effects of nanomaterials. J Appl Toxicol 2024; 44:1834-1853. [PMID: 38642013 DOI: 10.1002/jat.4609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/22/2024]
Abstract
Mitophagy is a selective cellular process critical for the removal of damaged mitochondria. It is essential in regulating mitochondrial number, ensuring mitochondrial functionality, and maintaining cellular equilibrium, ultimately influencing cell destiny. Numerous pathologies, such as neurodegenerative diseases, cardiovascular disorders, cancers, and various other conditions, are associated with mitochondrial dysfunctions. Thus, a detailed exploration of the regulatory mechanisms of mitophagy is pivotal for enhancing our understanding and for the discovery of novel preventive and therapeutic options for these diseases. Nanomaterials have become integral in biomedicine and various other sectors, offering advanced solutions for medical uses including biological imaging, drug delivery, and disease diagnostics and therapy. Mitophagy is vital in managing the cellular effects elicited by nanomaterials. This review provides a comprehensive analysis of the molecular mechanisms underpinning mitophagy, underscoring its significant influence on the biological responses of cells to nanomaterials. Nanoparticles can initiate mitophagy via various pathways, among which the PINK1-Parkin pathway is critical for cellular defense against nanomaterial-induced damage by promoting mitophagy. The role of mitophagy in biological effects was induced by nanomaterials, which are associated with alterations in Ca2+ levels, the production of reactive oxygen species, endoplasmic reticulum stress, and lysosomal damage.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Haitao Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
Torres M, Pederson B, Wang H, Lin LL, Wang HH, Bugarin-Lapuz A, Zhao Z, Qi L. Purkinje cell-specific deficiency in SEL1L-hrd1 endoplasmic reticulum-associated degradation causes progressive cerebellar ataxia in mice. JCI Insight 2024; 9:e174725. [PMID: 39352758 PMCID: PMC11563667 DOI: 10.1172/jci.insight.174725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Recent studies have identified multiple genetic variants of SEL1L-HRD1 endoplasmic reticulum-associated degradation (ERAD) in humans with neurodevelopmental disorders and locomotor dysfunctions, including ataxia. However, the relevance and importance of SEL1L-HRD1 ERAD in the pathogenesis of ataxia remain unexplored. Here, we showed that SEL1L deficiency in Purkinje cells leads to early-onset progressive cerebellar ataxia with progressive loss of Purkinje cells with age. Mice with Purkinje cell-specific deletion of SEL1L (Sel1LPcp2Cre) exhibited motor dysfunction beginning around 9 weeks of age. Transmission electron microscopy analysis revealed dilated ER and fragmented nuclei in Purkinje cells of adult Sel1LPcp2Cre mice, indicative of altered ER homeostasis and cell death. Finally, loss of Purkinje cells was associated with a secondary neurodegeneration of granular cells, as well as robust activation of astrocytes and proliferation of microglia, in the cerebellums of Sel1LPcp2Cre mice. These data demonstrate the pathophysiological importance of SEL1L-HRD1 ERAD in Purkinje cells in the pathogenesis of cerebellar ataxia.
Collapse
Affiliation(s)
- Mauricio Torres
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Brent Pederson
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hui Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Huilun Helen Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Amara Bugarin-Lapuz
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Wang L, Ren Z, Wu L, Zhang X, Wang M, Niu H, He X, Wang H, Chen Y, Shi G, Qian X. HRD1 reduction promotes cholesterol-induced vascular smooth muscle cell phenotypic change via endoplasmic reticulum stress. Mol Cell Biochem 2024; 479:3021-3036. [PMID: 38145449 DOI: 10.1007/s11010-023-04902-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/07/2023] [Indexed: 12/26/2023]
Abstract
Phenotypic change of vascular smooth muscle cells (VSMCs) is the main contributor of vascular pathological remodeling in atherosclerosis. The endoplasmic reticulum (ER) is critical for maintaining VSMC function through elimination of misfolded proteins that impair VSMC cellular function. ER-associated degradation (ERAD) is an ER-mediated process that controls protein quality by clearing misfolded proteins. One of the critical regulators of ERAD is HRD1, which also plays a vital role in lipid metabolism. However, the function of HRD1 in VSMCs of atherosclerotic vessels remains poorly understood. The level of HRD1 expression was analyzed in aortic tissues of mice fed with a high-fat diet (HFD). The H&E and EVG (VERHOEFF'S VAN GIESON) staining were used to demonstrate pathological vascular changes. IF (immunofluorescence) and WB (western blot) were used to explore the signaling pathways in vivo and in vitro. The wound closure and transwell assays were also used to test the migration rate of VSMCs. CRISPR gene editing and transcriptomic analysis were applied in vitro to explore the cellular mechanism. Our data showed significant reduction of HRD1 in aortic tissues of mice under HFD feeding. VSMC phenotypic change and HRD1 downregulation were detected by cholesterol supplement. Transcriptomic and further analysis of HRD1-KO VSMCs showed that HRD1 deficiency induced the expression of genes related to ER stress response, proliferation and migration, but reduced the contractile-related genes in VSMCs. HRD1 deficiency also exacerbated the proliferation, migration and ROS production of VSMCs induced by cholesterol, which promoted the VSMC dedifferentiation. Our results showed that HRD1 played an essential role in the contractile homeostasis of VSMCs by negatively regulating ER stress response. Thus, HRD1 in VSMCs could serve as a potential therapeutic target in metabolic disorder-induced vascular remodeling.
Collapse
Affiliation(s)
- Linli Wang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Zhitao Ren
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Lin Wu
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Ximei Zhang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Min Wang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Haiming Niu
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, 528400, China
| | - Xuemin He
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Heting Wang
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yanming Chen
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Guojun Shi
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Xiaoxian Qian
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
22
|
Song Z, Thepsuwan P, Hur WS, Torres M, Wu SA, Wei X, Tushi NJ, Wei J, Ferraresso F, Paton AW, Paton JC, Zheng Z, Zhang K, Fang D, Kastrup CJ, Jaiman S, Flick MJ, Sun S. Regulation of hepatic inclusions and fibrinogen biogenesis by SEL1L-HRD1 ERAD. Nat Commun 2024; 15:9244. [PMID: 39455574 PMCID: PMC11512042 DOI: 10.1038/s41467-024-53639-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Impaired secretion of an essential blood coagulation factor fibrinogen leads to hepatic fibrinogen storage disease (HFSD), characterized by the presence of fibrinogen-positive inclusion bodies and hypofibrinogenemia. However, the molecular mechanisms underlying the biogenesis of fibrinogen in the endoplasmic reticulum (ER) remain unexplored. Here we uncover a key role of SEL1L-HRD1 complex of ER-associated degradation (ERAD) in the formation of aberrant inclusion bodies, and the biogenesis of nascent fibrinogen protein complex in hepatocytes. Acute or chronic deficiency of SEL1L-HRD1 ERAD in the hepatocytes leads to the formation of hepatocellular inclusion bodies. Proteomics studies followed by biochemical assays reveal fibrinogen as a major component of the inclusion bodies. Mechanistically, we show that the degradation of misfolded endogenous fibrinogen Aα, Bβ, and γ chains by SEL1L-HRD1 ERAD is indispensable for the formation of a functional fibrinogen complex in the ER. Providing clinical relevance of these findings, SEL1L-HRD1 ERAD indeed degrades and thereby attenuates the pathogenicity of two disease-causing fibrinogen γ mutants. Together, this study demonstrates an essential role of SEL1L-HRD1 ERAD in fibrinogen biogenesis and provides insight into the pathogenesis of protein-misfolding diseases.
Collapse
Affiliation(s)
- Zhenfeng Song
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Pattaraporn Thepsuwan
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Woosuk Steve Hur
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Mauricio Torres
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Shuangcheng Alivia Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Nusrat Jahan Tushi
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Francesca Ferraresso
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Ze Zheng
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Christian J Kastrup
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sunil Jaiman
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Matthew James Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Shengyi Sun
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
23
|
Mao H, Kim GH, Pan L, Qi L. Regulation of leptin signaling and diet-induced obesity by SEL1L-HRD1 ER-associated degradation in POMC expressing neurons. Nat Commun 2024; 15:8435. [PMID: 39343970 PMCID: PMC11439921 DOI: 10.1038/s41467-024-52743-2] [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] [Received: 12/17/2023] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
Endoplasmic reticulum (ER) homeostasis in the hypothalamus has been implicated in the pathogenesis of diet-induced obesity (DIO) and type 2 diabetes; however, the underlying molecular mechanism remain vague and debatable. Here we report that SEL1L-HRD1 protein complex of the highly conserved ER-associated protein degradation (ERAD) machinery in POMC-expressing neurons ameliorates diet-induced obesity and its associated complications, partly by regulating the turnover of the long isoform of Leptin receptors (LepRb). Loss of SEL1L in POMC-expressing neurons attenuates leptin signaling and predisposes mice to HFD-associated pathologies including fatty liver, glucose intolerance, insulin and leptin resistance. Mechanistically, nascent LepRb, both wildtype and disease-associated Cys604Ser variant, are misfolding prone and bona fide substrates of SEL1L-HRD1 ERAD. In the absence of SEL1L-HRD1 ERAD, LepRb are largely retained in the ER, in an ER stress-independent manner. This study uncovers an important role of SEL1L-HRD1 ERAD in the pathogenesis of central leptin resistance and leptin signaling.
Collapse
Affiliation(s)
- Hancheng Mao
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Geun Hyang Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York, NY, 10591, USA
| | - Linxiu Pan
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Ling Qi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
24
|
Guo M, Liu R, Zhang F, Qu J, Yang Y, Li X. A new perspective on liver diseases: Focusing on the mitochondria-associated endoplasmic reticulum membranes. Pharmacol Res 2024; 208:107409. [PMID: 39284429 DOI: 10.1016/j.phrs.2024.107409] [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: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The pathogenesis of liver diseases is multifaceted and intricate, posing a persistent global public health challenge with limited therapeutic options. Therefore, further research into liver diseases is imperative for better comprehension and advancement in treatment strategies. Numerous studies have confirmed the endoplasmic reticulum (ER) and mitochondria as key organelles driving liver diseases. Notably, the mitochondrial-associated ER membranes (MAMs) establish a physical and functional connection between the ER and mitochondria, highlighting the importance of inter-organelle communication in maintaining their functional homeostasis. This review delves into the intricate architecture and regulative mechanism of the integrated MAM that facilitate the physiological transfer of signals and substances between organelles. Additionally, we also provide a detailed overview regarding the varied pathogenic roles of malfunctioning MAM in liver diseases, focusing on its involvement in the progression of ER stress and mitochondrial dysfunction, the regulation of mitochondrial dynamics and Ca2+ transfer, as well as the disruption of lipid and glucose homeostasis. Furthermore, the current challenges and prospects associated with MAM in liver disease research are thoroughly discussed. In conclusion, elucidating the specific structure and function of MAM in different liver diseases may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Mengyu Guo
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 100029, China
| | - Fukun Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China.
| |
Collapse
|
25
|
Yan Y, Huang J, Chen X, Li Y, Zhao W, Li C. UFL1 regulates cellular homeostasis by targeting endoplasmic reticulum and mitochondria in NEFA-stimulated bovine mammary epithelial cells via the IRE1α/XBP1 pathway. Free Radic Biol Med 2024; 222:16-26. [PMID: 38821134 DOI: 10.1016/j.freeradbiomed.2024.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Elevated levels of NEFA caused by negative energy balance in transition cows induce cellular dyshomeostasis. Ubiquitin-like modifier 1 ligating enzyme 1 (UFL1) can maintain cellular homeostasis and act as a critical regulator of stress responses besides functioning in the ubiquitin-like system. The objective of this study was to elucidate the UFL1 working mechanism on promoting cellular adaptations in bovine mammary epithelial cells (BMECs) in response to NEFA challenge, with an emphasis on the ER and mitochondrial function. The results showed that exogenous NEFA and UFL1 depletion resulted in the disorder of ER and mitochondrial homeostasis and the damage of BMEC integrity, overexpression of UFL1 effectively alleviated the NEFA-induced cellular dyshomeostasis. Mechanistically, our study found that UFL1 had a strong interaction with IRE1α and could modulate the IRE1α/XBP1 pathway of unfolded protein response in NEFA-stimulated BMECs, thereby contributing to the modulation of cellular homeostasis. These findings imply that targeting UFL1 may be a therapeutic alternative to relieve NEB-induced metabolic changes in perinatal dairy cows.
Collapse
Affiliation(s)
- Yexiao Yan
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Junpeng Huang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Xiangxing Chen
- Zibo Service Center for Animal Husbandry and Fishery, Zibo, 255000, China
| | - Yuan Li
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Weiguo Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Chengmin Li
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| |
Collapse
|
26
|
Knupp J, Chen YJ, Wang E, Arvan P, Tsai B. Sigma-1 receptor recruits LC3 mRNA to ER-associated omegasomes to promote localized LC3 translation enabling functional autophagy. Cell Rep 2024; 43:114619. [PMID: 39128005 PMCID: PMC11376464 DOI: 10.1016/j.celrep.2024.114619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/14/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Autophagosome formation initiated on the endoplasmic reticulum (ER)-associated omegasome requires LC3. Translational regulation of LC3 biosynthesis is unexplored. Here we demonstrate that LC3 mRNA is recruited to omegasomes by directly binding to the ER transmembrane Sigma-1 receptor (S1R). Cell-based and in vitro reconstitution experiments show that S1R interacts with the 3' UTR of LC3 mRNA and ribosomes to promote LC3 translation. Strikingly, the 3' UTR of LC3 is also required for LC3 protein lipidation, thereby linking the mRNA-3' UTR to LC3 function. An autophagy-defective S1R mutant responsible for amyotrophic lateral sclerosis cannot bind LC3 mRNA or induce LC3 translation. We propose a model wherein S1R de-represses LC3 mRNA via its 3' UTR at the ER, enabling LC3 biosynthesis and lipidation. Because several other LC3-related proteins use the same mechanism, our data reveal a conserved pathway for localized translation essential for autophagosome biogenesis with insights illuminating the molecular basis of a neurodegenerative disease.
Collapse
Affiliation(s)
- Jeffrey Knupp
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, 1135 Catherine Street, Ann Arbor, MI 48109 USA
| | - Yu-Jie Chen
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA
| | - Emily Wang
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA
| | - Peter Arvan
- Cellular and Molecular Biology Program, University of Michigan Medical School, 1135 Catherine Street, Ann Arbor, MI 48109 USA; Division of Metabolism Endocrinology & Diabetes, University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA.
| | - Billy Tsai
- Department of Cell & Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3043, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, 1135 Catherine Street, Ann Arbor, MI 48109 USA.
| |
Collapse
|
27
|
Wang J, Fan P, Shen P, Fan C, Zhao P, Yao Shen, Dong K, Ling R, Chen S, Zhang J. XBP1s activates METTL3/METTL14 for ER-phagy and paclitaxel sensitivity regulation in breast cancer. Cancer Lett 2024; 596:216846. [PMID: 38582397 DOI: 10.1016/j.canlet.2024.216846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/25/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
Cancer cells employ the unfolded protein response (UPR) or induce autophagy, especially selective removal of certain ER domains via reticulophagy (termed ER-phagy), to mitigate endoplasmic reticulum (ER) stress for ER homeostasis when encountering microenvironmental stress. N6-methyladenosine (m6A) is one of the most abundant epitranscriptional modifications and plays important roles in various biological processes. However, the molecular mechanism of m6A modification in the ER stress response is poorly understood. In this study, we first found that ER stress could dramatically elevate m6A methylation levels through XBP1s-dependent transcriptional upregulation of METTL3/METTL14 in breast cancer (BC) cells. Further MeRIP sequencing and relevant validation results confirmed that ER stress caused m6A methylation enrichment on target genes for ER-phagy. Mechanistically, METTL3/METTL14 increased ER-phagy machinery formation by promoting m6A modification of the ER-phagy regulators CALCOCO1 and p62, thus enhancing their mRNA stability. Of note, we further confirmed that the chemotherapeutic drug paclitaxel (PTX) could induce ER stress and increase m6A methylation for ER-phagy. Furthermore, the combination of METTL3/METTL14 inhibitors with PTX demonstrated a significant synergistic therapeutic effect in both BC cells and xenograft mice. Thus, our data built a novel bridge on the crosstalk between ER stress, m6A methylation and ER-phagy. Most importantly, our work provides novel evidence of METTL3 and METTL14 as potential therapeutic targets for PTX sensitization in breast cancer.
Collapse
Affiliation(s)
- Jiajia Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Pengyu Fan
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Peng Shen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Cong Fan
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Pan Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yao Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Kewei Dong
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Ling
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Suning Chen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Jian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
28
|
Wu S, Liu P, Cvetanovic M, Lin W. Endoplasmic reticulum associated degradation preserves neurons viability by maintaining endoplasmic reticulum homeostasis. Front Neurosci 2024; 18:1437854. [PMID: 39135735 PMCID: PMC11317260 DOI: 10.3389/fnins.2024.1437854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) is a principal quality-control mechanism responsible for targeting misfolded ER proteins for cytosolic degradation. Evidence suggests that impairment of ERAD contributes to neuron dysfunction and death in neurodegenerative diseases, many of which are characterized by accumulation and aggregation of misfolded proteins. However, the physiological role of ERAD in neurons remains unclear. The Sel1L-Hrd1 complex consisting of the E3 ubiquitin ligase Hrd1 and its adaptor protein Sel1L is the best-characterized ERAD machinery. Herein, we showed that Sel1L deficiency specifically in neurons of adult mice impaired the ERAD activity of the Sel1L-Hrd1 complex and led to disruption of ER homeostasis, ER stress and activation of the unfold protein response (UPR). Adult mice with Sel1L deficiency in neurons exhibited weight loss and severe motor dysfunction, and rapidly succumbed to death. Interestingly, Sel1L deficiency in neurons caused global brain atrophy, particularly cerebellar and hippocampal atrophy, in adult mice. Moreover, we found that cerebellar and hippocampal atrophy in these mice resulted from degeneration of Purkinje neurons and hippocampal neurons, respectively. These findings indicate that ERAD is required for maintaining ER homeostasis and the viability and function of neurons in adults under physiological conditions.
Collapse
Affiliation(s)
- Shuangchan Wu
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Pingting Liu
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
29
|
Qian B, Li TY, Zheng ZX, Zhang HY, Xu WQ, Mo SM, Cui JJ, Chen WJ, Lin YC, Lin ZN. The involvement of SigmaR1 K142 degradation mediated by ERAD in neural senescence linked with CdCl 2 exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134466. [PMID: 38718507 DOI: 10.1016/j.jhazmat.2024.134466] [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: 12/20/2023] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/30/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Due to its uncertain pathogenesis, there is currently no treatment available for AD. Increasing evidences have linked cellular senescence to AD, although the mechanism triggering cellular senescence in AD requires further exploration. To investigate the involvement of cellular senescence in AD, we explored the effects of cadmium chloride (CdCl2) exposure, one of the potential environmental risk factors for AD, on neuron senescence in vivo and in vitro. β-amyloid (Aβ) and tubulin-associated protein (tau) pathologies were found to be enhanced by CdCl2 exposure in the in vitro models, while p53/p21/Rb cascade-related neuronal senescence pathways were activated. Conversely, the use of melatonin, a cellular senescence inhibitor, or a cadmium ion chelator suppressed CdCl2-induced neuron senescence, along with the Aβ and tau pathologies. Mechanistically, CdCl2 exposure activated the suppressor enhancer Lin-12/Notch 1-like (SEL1L)/HMG-CoA reductase degradation 1 (HRD1)-regulated endoplasmic reticulum-associated degradation (ERAD), which enhanced the ubiquitin degradation of sigma-1 receptor (SigmaR1) by specifically recognizing its K142 site, resulting in the activation of the p53/p21/Rb pathway via the induction of Ca2+ dyshomeostasis and mitochondrial dysfunction. In the in vivo models, the administration of the SigmaR1 agonist ANAVEX2-73 rescues neurobehavioral inhibition and alleviates cellular senescence and AD-like pathology in the brain tissue of CdCl2-exposed mice. Consequently, the present study revealed a novel senescence-associated regulatory route for the SEL1L/HRD1/SigmaR1 axis that affects the pathological progression of CdCl2 exposure-associated AD. CdCl2 exposure activated SEL1L/HRD1-mediated ERAD and promoted the ubiquitinated degradation of SigmaR1, activating p53/p21/Rb pathway-regulated neuronal senescence. The results of the present study suggest that SigmaR1 may function as a neuroprotective biomarker of neuronal senescence, and pharmacological activation of SigmaR1 could be a promising intervention strategy for AD therapy.
Collapse
Affiliation(s)
- Bo Qian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ting-Yu Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhao-Xuan Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Han-Yu Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wen-Qi Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Su-Min Mo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jia-Jia Cui
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei-Jie Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yu-Chun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhong-Ning Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
30
|
Mu W, Zhi Y, Zhou J, Wang C, Chai K, Fan Z, Lv G. Endoplasmic reticulum stress and quality control in relation to cisplatin resistance in tumor cells. Front Pharmacol 2024; 15:1419468. [PMID: 38948460 PMCID: PMC11211601 DOI: 10.3389/fphar.2024.1419468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
The endoplasmic reticulum (ER) is a crucial organelle that orchestrates key cellular functions like protein folding and lipid biosynthesis. However, it is highly sensitive to disturbances that lead to ER stress. In response, the unfolded protein response (UPR) activates to restore ER homeostasis, primarily through three sensors: IRE1, ATF6, and PERK. ERAD and autophagy are crucial in mitigating ER stress, yet their dysregulation can lead to the accumulation of misfolded proteins. Cisplatin, a commonly used chemotherapy drug, induces ER stress in tumor cells, activating complex signaling pathways. Resistance to cisplatin stems from reduced drug accumulation, activation of DNA repair, and anti-apoptotic mechanisms. Notably, cisplatin-induced ER stress can dualistically affect tumor cells, promoting either survival or apoptosis, depending on the context. ERAD is crucial for degrading misfolded proteins, whereas autophagy can protect cells from apoptosis or enhance ER stress-induced apoptosis. The complex interaction between ER stress, cisplatin resistance, ERAD, and autophagy opens new avenues for cancer treatment. Understanding these processes could lead to innovative strategies that overcome chemoresistance, potentially improving outcomes of cisplatin-based cancer treatments. This comprehensive review provides a multifaceted perspective on the complex mechanisms of ER stress, cisplatin resistance, and their implications in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
31
|
Correa-Medero LO, Jankowski SE, Hong HS, Armas ND, Vijendra AI, Reynolds MB, Fogo GM, Awad D, Dils AT, Inoki KA, Williams RG, Ye AM, Svezhova N, Gomez-Rivera F, Collins KL, O'Riordan MX, Sanderson TH, Lyssiotis CA, Carty SA. ER-associated degradation adapter Sel1L is required for CD8 + T cell function and memory formation following acute viral infection. Cell Rep 2024; 43:114156. [PMID: 38687642 PMCID: PMC11194752 DOI: 10.1016/j.celrep.2024.114156] [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: 10/09/2023] [Revised: 03/06/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
The maintenance of antigen-specific CD8+ T cells underlies the efficacy of vaccines and immunotherapies. Pathways contributing to CD8+ T cell loss are not completely understood. Uncovering the pathways underlying the limited persistence of CD8+ T cells would be of significant benefit for developing novel strategies of promoting T cell persistence. Here, we demonstrate that murine CD8+ T cells experience endoplasmic reticulum (ER) stress following activation and that the ER-associated degradation (ERAD) adapter Sel1L is induced in activated CD8+ T cells. Sel1L loss limits CD8+ T cell function and memory formation following acute viral infection. Mechanistically, Sel1L is required for optimal bioenergetics and c-Myc expression. Finally, we demonstrate that human CD8+ T cells experience ER stress upon activation and that ER stress is negatively associated with improved T cell functionality in T cell-redirecting therapies. Together, these results demonstrate that ER stress and ERAD are important regulators of T cell function and persistence.
Collapse
Affiliation(s)
- Luis O Correa-Medero
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Hanna S Hong
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas D Armas
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Mack B Reynolds
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Garrett M Fogo
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dominik Awad
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexander T Dils
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Reid G Williams
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Nadezhda Svezhova
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Kathleen L Collins
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas H Sanderson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shannon A Carty
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
Lam CHI, Zuo B, Chan HHL, Leung TW, Abokyi S, Catral KPC, Tse DYY. Coenzyme Q10 eyedrops conjugated with vitamin E TPGS alleviate neurodegeneration and mitochondrial dysfunction in the diabetic mouse retina. Front Cell Neurosci 2024; 18:1404987. [PMID: 38863499 PMCID: PMC11165046 DOI: 10.3389/fncel.2024.1404987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness and vision impairment worldwide and represents one of the most common complications among diabetic patients. Current treatment modalities for DR, including laser photocoagulation, intravitreal injection of corticosteroid, and anti-vascular endothelial growth factor (VEGF) agents, target primarily vascular lesions. However, these approaches are invasive and have several limitations, such as potential loss of visual function, retinal scars and cataract formation, and increased risk of ocular hypertension, vitreous hemorrhage, retinal detachment, and intraocular inflammation. Recent studies have suggested mitochondrial dysfunction as a pivotal factor leading to both the vascular and neural damage in DR. Given that Coenzyme Q10 (CoQ10) is a proven mitochondrial stabilizer with antioxidative properties, this study investigated the effect of CoQ10 eyedrops [in conjunction with vitamin E d-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS)] on DR-induced neurodegeneration using a type 2 diabetes mouse model (C57BLKsJ-db/db mice). Utilizing a comprehensive electroretinography protocol, supported by immunohistochemistry, our results revealed that topical application of CoQ10 eyedrops conjugated with vitamin E TPGS produced a neuroprotective effect against diabetic-induced neurodegeneration by preserving the function and histology of various retinal neural cell types. Compared to the control group, mice treated with CoQ10 exhibited thicker outer and inner nuclear layers, higher densities of photoreceptor, cone cell, and rod-bipolar cell dendritic boutons, and reduced glial reactivity and microglial cell density. Additionally, the CoQ10 treatment significantly alleviated retinal levels of MMP-9 and enhanced mitochondrial function. These findings provide further insight into the role of mitochondrial dysfunction in the development of DR and suggest CoQ10 eyedrops, conjugated with vitamin E TPGS, as a potential complementary therapy for DR-related neuropathy.
Collapse
Affiliation(s)
- Christie Hang-I Lam
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Centre for Eye and Vision Research Limited, Shatin, Hong Kong SAR, China
| | - Bing Zuo
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Henry Ho-Lung Chan
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Centre for Eye and Vision Research Limited, Shatin, Hong Kong SAR, China
| | - Tsz-Wing Leung
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Samuel Abokyi
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | | | - Dennis Yan-Yin Tse
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Centre for Eye and Vision Research Limited, Shatin, Hong Kong SAR, China
- Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
33
|
Hinton AO, N'jai AU, Vue Z, Wanjalla C. Connection Between HIV and Mitochondria in Cardiovascular Disease and Implications for Treatments. Circ Res 2024; 134:1581-1606. [PMID: 38781302 PMCID: PMC11122810 DOI: 10.1161/circresaha.124.324296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
HIV infection and antiretroviral therapy alter mitochondrial function, which can progressively lead to mitochondrial damage and accelerated aging. The interaction between persistent HIV reservoirs and mitochondria may provide insight into the relatively high rates of cardiovascular disease and mortality in persons living with HIV. In this review, we explore the intricate relationship between HIV and mitochondrial function, highlighting the potential for novel therapeutic strategies in the context of cardiovascular diseases. We reflect on mitochondrial dynamics, mitochondrial DNA, and mitochondrial antiviral signaling protein in the context of HIV. Furthermore, we summarize how toxicities related to early antiretroviral therapy and current highly active antiretroviral therapy can contribute to mitochondrial dysregulation, chronic inflammation, and poor clinical outcomes. There is a need to understand the mechanisms and develop new targeted therapies. We further consider current and potential future therapies for HIV and their interplay with mitochondria. We reflect on the next-generation antiretroviral therapies and HIV cure due to the direct and indirect effects of HIV persistence, associated comorbidities, coinfections, and the advancement of interdisciplinary research fields. This includes exploring novel and creative approaches to target mitochondria for therapeutic intervention.
Collapse
Affiliation(s)
- Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Alhaji U N'jai
- Biological Sciences, Fourah Bay College and College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone and Koinadugu College, Kabala (A.U.N.)
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Celestine Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.W.)
| |
Collapse
|
34
|
Wu Z, Xiao C, Li F, Huang W, You F, Li X. Mitochondrial fusion-fission dynamics and its involvement in colorectal cancer. Mol Oncol 2024; 18:1058-1075. [PMID: 38158734 PMCID: PMC11076987 DOI: 10.1002/1878-0261.13578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/21/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
The incidence and mortality rates of colorectal cancer have elevated its status as a significant public health concern. Recent research has elucidated the crucial role of mitochondrial fusion-fission dynamics in the initiation and progression of colorectal cancer. Elevated mitochondrial fission or fusion activity can contribute to the metabolic reprogramming of tumor cells, thereby activating oncogenic pathways that drive cell proliferation, invasion, migration, and drug resistance. Nevertheless, excessive mitochondrial fission can induce apoptosis, whereas moderate mitochondrial fusion can protect cells from oxidative stress. This imbalance in mitochondrial dynamics can exert dual roles as both promoters and inhibitors of colorectal cancer progression. This review provides an in-depth analysis of the fusion-fission dynamics and the underlying pathological mechanisms in colorectal cancer cells. Additionally, it offers partial insights into the mitochondrial kinetics in colorectal cancer-associated cells, such as immune and endothelial cells. This review is aimed at identifying key molecular events involved in colorectal cancer progression and highlighting the potential of mitochondrial dynamic proteins as emerging targets for pharmacological intervention.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Oncology Teaching and Research DepartmentChengdu University of Traditional Chinese MedicineChina
| | - Fang Li
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Institute of OncologyChengdu University of Traditional Chinese MedicineChina
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Oncology Teaching and Research DepartmentChengdu University of Traditional Chinese MedicineChina
| |
Collapse
|
35
|
Li D, Rocha-Roa C, Schilling MA, Reinisch KM, Vanni S. Lipid scrambling is a general feature of protein insertases. Proc Natl Acad Sci U S A 2024; 121:e2319476121. [PMID: 38621120 PMCID: PMC11047089 DOI: 10.1073/pnas.2319476121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/13/2024] [Indexed: 04/17/2024] Open
Abstract
Glycerophospholipids are synthesized primarily in the cytosolic leaflet of the endoplasmic reticulum (ER) membrane and must be equilibrated between bilayer leaflets to allow the ER and membranes derived from it to grow. Lipid equilibration is facilitated by integral membrane proteins called "scramblases." These proteins feature a hydrophilic groove allowing the polar heads of lipids to traverse the hydrophobic membrane interior, similar to a credit card moving through a reader. Nevertheless, despite their fundamental role in membrane expansion and dynamics, the identity of most scramblases has remained elusive. Here, combining biochemical reconstitution and molecular dynamics simulations, we show that lipid scrambling is a general feature of protein insertases, integral membrane proteins which insert polypeptide chains into membranes of the ER and organelles disconnected from vesicle trafficking. Our data indicate that lipid scrambling occurs in the same hydrophilic channel through which protein insertion takes place and that scrambling is abolished in the presence of nascent polypeptide chains. We propose that protein insertases could have a so-far-overlooked role in membrane dynamics as scramblases.
Collapse
Affiliation(s)
- Dazhi Li
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Cristian Rocha-Roa
- Department of Biology, University of Fribourg, FribourgCH-1700, Switzerland
| | - Matthew A. Schilling
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06520
| | - Stefano Vanni
- Department of Biology, University of Fribourg, FribourgCH-1700, Switzerland
- Swiss National Center for Competence in Research Bio-Inspired Materials, University of Fribourg, FribourgCH-1700, Switzerland
| |
Collapse
|
36
|
Lin H, Guo X, Liu J, Tang Y, Chen L, Chen H, Zhao Y, Wang L, Li H, Yu J, Yao P. Ethanol-Induced Hepatic Ferroptosis Is Mediated by PERK-Dependent MAMs Formation: Preventive Role of Quercetin. Mol Nutr Food Res 2024; 68:e2300343. [PMID: 38501770 DOI: 10.1002/mnfr.202300343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/05/2023] [Indexed: 03/20/2024]
Abstract
SCOPE Iron deposition is frequently observed in alcoholic liver disease (ALD), which indicates a potential role of ferroptosis in its development. This study aims to explore the effects of quercetin on ferroptosis in ALD and elucidates the underlying mechanism involving the formation of mitochondria-associated endoplasmic reticulum membranes (MAMs) mediated by protein kinase RNA-like endoplasmic reticulum kinase (PERK). METHODS AND RESULTS C57BL/6J mice are fed either a regular or an ethanol-containing liquid diet (with 28% energy form ethanol) with or without quercetin supplementation (100 mg kg-1 BW) for 12 weeks. Ethanol feeding or treatment induced ferroptosis in mice and AML12 cells, which is associated with increased MAMs formation and PERK expression within MAMs. Quercetin attenuates these changes and protects against ethanol-induced liver injury. The antiferroptotic effect of quercetin is abolished by ferroptosis inducers, but mimicked by ferroptosis inhibitors and PERK knockdown. The study demonstrates that PERK structure, rather than its kinase activity (transfected with the K618A site mutation that inhibits kinase activity-ΔK plasmid or protein C terminal knockout-ΔC plasmid of PERK), mediates the enhanced MAMs formation and ferroptosis during the ethanol exposure. CONCLUSION Quercetin ameliorates ethanol-induced liver injury by inhibiting ferroptosis via modulating PERK-dependent MAMs formation.
Collapse
Affiliation(s)
- Hongkun Lin
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
- Department of Nutrition, School of Public Health, Wuhan University, Wuhan, 430071, P. R. China
| | - Xiaoping Guo
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Jingjing Liu
- Henan Provincial Center for Disease Control and Prevention, Zhengzhou, 450016, P. R. China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Li Chen
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Huimin Chen
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Ying Zhao
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Lili Wang
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Hongxia Li
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| | - Jiasheng Yu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health,Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
- Ministry of Education Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan, 430030, P. R. China
| |
Collapse
|
37
|
Jenkins BC, Neikirk K, Katti P, Claypool SM, Kirabo A, McReynolds MR, Hinton A. Mitochondria in disease: changes in shapes and dynamics. Trends Biochem Sci 2024; 49:346-360. [PMID: 38402097 PMCID: PMC10997448 DOI: 10.1016/j.tibs.2024.01.011] [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: 09/05/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 02/26/2024]
Abstract
Mitochondrial structure often determines the function of these highly dynamic, multifunctional, eukaryotic organelles, which are essential for maintaining cellular health. The dynamic nature of mitochondria is apparent in descriptions of different mitochondrial shapes [e.g., donuts, megamitochondria (MGs), and nanotunnels] and crista dynamics. This review explores the significance of dynamic alterations in mitochondrial morphology and regulators of mitochondrial and cristae shape. We focus on studies across tissue types and also describe new microscopy techniques for detecting mitochondrial morphologies both in vivo and in vitro that can improve understanding of mitochondrial structure. We highlight the potential therapeutic benefits of regulating mitochondrial morphology and discuss prospective avenues to restore mitochondrial bioenergetics to manage diseases related to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Brenita C Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Steven M Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801, USA.
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
38
|
Li X, Yang JY, Hu WZ, Ruan Y, Chen HY, Zhang Q, Zhang Z, Ding ZS. Mitochondria-associated membranes contribution to exercise-mediated alleviation of hepatic insulin resistance: Contrasting high-intensity interval training with moderate-intensity continuous training in a high-fat diet mouse model. J Diabetes 2024; 16:e13540. [PMID: 38599845 PMCID: PMC11006604 DOI: 10.1111/1753-0407.13540] [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: 07/06/2023] [Revised: 12/18/2023] [Accepted: 02/03/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVE Mitochondria-associated membranes (MAMs) serve pivotal functions in hepatic insulin resistance (IR). Our aim was to explore the potential role of MAMs in mitigating hepatic IR through exercise and to compare the effects of different intensities of exercise on hepatic MAMs formation in high-fat diet (HFD) mice. METHODS Male C57BL/6J mice were fed an HFD and randomly assigned to undergo supervised high-intensity interval training (HIIT) or moderate-intensity continuous training (MICT). IR was evaluated using the serum triglyceride/high-density lipoprotein cholesterol ratio (TG/HDL-C), glucose tolerance test (GTT), and insulin tolerance test (ITT). Hepatic steatosis was observed using hematoxylin-eosin (H&E) and oil red O staining. The phosphatidylinositol 3-kinase/protein kinase B/glycogen synthase kinase 3 beta (PI3K-AKT-GSK3β) signaling pathway was assessed to determine hepatic IR. MAMs were evaluated through immunofluorescence (colocalization of voltage-dependent anion-selective channel 1 [VDAC1] and inositol 1,4,5-triphosphate receptor [IP3R]). RESULTS After 8 weeks on an HFD, there was notable inhibition of the hepatic PI3K/Akt/GSK3β signaling pathway, accompanied by a marked reduction in hepatic IP3R-VDAC1 colocalization levels. Both 8-week HIIT and MICT significantly enhanced the hepatic PI3K/Akt/GSK3β signaling and colocalization levels of IP3R-VDAC1 in HFD mice, with MICT exhibiting a stronger effect on hepatic MAMs formation. Furthermore, the colocalization of hepatic IP3R-VDAC1 positively correlated with the expression levels of phosphorylation of protein kinase B (p-AKT) and phosphorylation of glycogen synthase kinase 3 beta (p-GSK3β), while displaying a negative correlation with serum triglyceride/high-density lipoprotein cholesterol levels. CONCLUSION The reduction in hepatic MAMs formation induced by HFD correlates with the development of hepatic IR. Both HIIT and MICT effectively bolster hepatic MAMs formation in HFD mice, with MICT demonstrating superior efficacy. Thus, MAMs might wield a pivotal role in exercise-induced alleviation of hepatic IR.
Collapse
Affiliation(s)
- Xi Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - Jun Yang Yang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - Wen Zhi Hu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - YuXin Ruan
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - Hong Ying Chen
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - Qiang Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - Zhe Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| | - Zhe Shu Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of EducationEast China Normal UniversityShanghaiChina
- College of Physical Education & HealthEast China Normal UniversityShanghaiChina
| |
Collapse
|
39
|
Liu Y, Huo JL, Ren K, Pan S, Liu H, Zheng Y, Chen J, Qiao Y, Yang Y, Feng Q. Mitochondria-associated endoplasmic reticulum membrane (MAM): a dark horse for diabetic cardiomyopathy treatment. Cell Death Discov 2024; 10:148. [PMID: 38509100 PMCID: PMC10954771 DOI: 10.1038/s41420-024-01918-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/25/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024] Open
Abstract
Diabetic cardiomyopathy (DCM), an important complication of diabetes mellitus (DM), is one of the most serious chronic heart diseases and has become a major cause of heart failure worldwide. At present, the pathogenesis of DCM is unclear, and there is still a lack of effective therapeutics. Previous studies have shown that the homeostasis of mitochondria and the endoplasmic reticulum (ER) play a core role in maintaining cardiovascular function, and structural and functional abnormalities in these organelles seriously impact the occurrence and development of various cardiovascular diseases, including DCM. The interplay between mitochondria and the ER is mediated by the mitochondria-associated ER membrane (MAM), which participates in regulating energy metabolism, calcium homeostasis, mitochondrial dynamics, autophagy, ER stress, inflammation, and other cellular processes. Recent studies have proven that MAM is closely related to the initiation and progression of DCM. In this study, we aim to summarize the recent research progress on MAM, elaborate on the key role of MAM in DCM, and discuss the potential of MAM as an important therapeutic target for DCM, thereby providing a theoretical reference for basic and clinical studies of DCM treatment.
Collapse
Affiliation(s)
- Yong Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Henan Province Research Center for Kidney Disease, 450052, Zhengzhou, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, 450052, Zhengzhou, P. R. China
| | - Jin-Ling Huo
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Henan Province Research Center for Kidney Disease, 450052, Zhengzhou, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, 450052, Zhengzhou, P. R. China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Shaokang Pan
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Henan Province Research Center for Kidney Disease, 450052, Zhengzhou, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, 450052, Zhengzhou, P. R. China
| | - Hengdao Liu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Yifeng Zheng
- Institute for Biomedical Sciences, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano, 399-4598, Japan
| | - Jingfang Chen
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- Henan Province Research Center for Kidney Disease, 450052, Zhengzhou, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, 450052, Zhengzhou, P. R. China
| | - Yingjin Qiao
- Blood Purification Center, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
| | - Yang Yang
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
| | - Qi Feng
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China.
- Henan Province Research Center for Kidney Disease, 450052, Zhengzhou, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, 450052, Zhengzhou, P. R. China.
| |
Collapse
|
40
|
Cao H, Zhou X, Xu B, Hu H, Guo J, Ma Y, Wang M, Li N, Jun Z. Advances in the study of protein folding and endoplasmic reticulum-associated degradation in mammal cells. J Zhejiang Univ Sci B 2024; 25:212-232. [PMID: 38453636 PMCID: PMC10918413 DOI: 10.1631/jzus.b2300403] [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: 06/11/2023] [Accepted: 08/03/2023] [Indexed: 03/09/2024]
Abstract
The endoplasmic reticulum is a key site for protein production and quality control. More than one-third of proteins are synthesized and folded into the correct three-dimensional conformation in the endoplasmic reticulum. However, during protein folding, unfolded and/or misfolded proteins are prone to occur, which may lead to endoplasmic reticulum stress. Organisms can monitor the quality of the proteins produced by endoplasmic reticulum quality control (ERQC) and endoplasmic reticulum-associated degradation (ERAD), which maintain endoplasmic reticulum protein homeostasis by degrading abnormally folded proteins. The underlying mechanisms of protein folding and ERAD in mammals have not yet been fully explored. Therefore, this paper reviews the process and function of protein folding and ERAD in mammalian cells, in order to help clinicians better understand the mechanism of ERAD and to provide a scientific reference for the treatment of diseases caused by abnormal ERAD.
Collapse
Affiliation(s)
- Hong Cao
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Xuchang Zhou
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Bowen Xu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Han Hu
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China
| | - Jianming Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Yuwei Ma
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Miao Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China
| | - Nan Li
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai 200433, China.
| | - Zou Jun
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
41
|
Pan X, Ren L, Yang Y, Xu Y, Ning L, Zhang Y, Luo H, Zou Q, Zhang Y. MCSdb, a database of proteins residing in membrane contact sites. Sci Data 2024; 11:281. [PMID: 38459036 PMCID: PMC10923927 DOI: 10.1038/s41597-024-03104-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 02/29/2024] [Indexed: 03/10/2024] Open
Abstract
Organelles do not act as autonomous discrete units but rather as interconnected hubs that engage in extensive communication by forming close contacts called "membrane contact sites (MCSs)". And many proteins have been identified as residing in MCS and playing important roles in maintaining and fulfilling specific functions within these microdomains. However, a comprehensive compilation of these MCS proteins is still lacking. Therefore, we developed MCSdb, a manually curated resource of MCS proteins and complexes from publications. MCSdb documents 7010 MCS protein entries and 263 complexes, involving 24 organelles and 44 MCSs across 11 species. Additionally, MCSdb orchestrates all data into different categories with multitudinous information for presenting MCS proteins. In summary, MCSdb provides a valuable resource for accelerating MCS functional interpretation and interorganelle communication deciphering.
Collapse
Affiliation(s)
- Xianrun Pan
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liping Ren
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Yu Yang
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
| | - Yi Xu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Ning
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, China
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yibing Zhang
- Glasgow College, University of Electronic Science and Technology of China, Chengdu, China
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| | - Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
42
|
Podolsky MJ, Kheyfets B, Pandey M, Beigh AH, Yang CD, Lizama CO, Datta R, Lin LL, Wang Z, Wolters PJ, McManus MT, Qi L, Atabai K. Genome-wide screens identify SEL1L as an intracellular rheostat controlling collagen turnover. Nat Commun 2024; 15:1531. [PMID: 38378719 PMCID: PMC10879544 DOI: 10.1038/s41467-024-45817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 01/30/2024] [Indexed: 02/22/2024] Open
Abstract
Accumulating evidence has implicated impaired extracellular matrix (ECM) clearance as a key factor in fibrotic disease. Despite decades of research elucidating the effectors of ECM clearance, relatively little is understood regarding the upstream regulation of this process. Collagen is the most abundant constituent of normal and fibrotic ECM in mammalian tissues. Its catabolism occurs through extracellular proteolysis and cell-mediated uptake of collagen fragments for intracellular degradation. Given the paucity of information regarding the regulation of this latter process, here we execute unbiased genome-wide screens to understand the molecular underpinnings of cell-mediated collagen clearance. Using this approach, we discover a mechanism through which collagen biosynthesis is sensed by cells internally and directly regulates clearance of extracellular collagen. The sensing mechanism appears to be dependent on endoplasmic reticulum-resident protein SEL1L and occurs via a noncanonical function of this protein. This pathway functions as a homeostatic negative feedback loop that limits collagen accumulation in tissues. In human fibrotic lung disease, the induction of this collagen clearance pathway by collagen synthesis is impaired, thereby contributing to the pathological accumulation of collagen in lung tissue. Thus, we describe cell-autonomous, rheostatic collagen clearance as an important pathway of tissue homeostasis.
Collapse
Affiliation(s)
- Michael J Podolsky
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Benjamin Kheyfets
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Monika Pandey
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Afaq H Beigh
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Christopher D Yang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Ritwik Datta
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Liangguang L Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Zhihong Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Michael T McManus
- Department of Microbiology and Immunology and UCSF Diabetes Center, University of California, San Francisco, CA, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, CA, USA.
- Lung Biology Center, University of California, San Francisco, CA, USA.
| |
Collapse
|
43
|
Zhao X, Liu D, Zhao Y, Wang Z, Wang Y, Chen Z, Ning S, Wang G, Meng L, Yao J, Tian X. HRD1-induced TMEM2 ubiquitination promotes ER stress-mediated apoptosis through a non-canonical pathway in intestinal ischemia/reperfusion. Cell Death Dis 2024; 15:154. [PMID: 38378757 PMCID: PMC10879504 DOI: 10.1038/s41419-024-06504-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/22/2024]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a typical pathological course in the clinic with a high morbidity rate. Recent research has pointed out the critical role of ubiquitination during the occurrence and development of intestinal I/R by precisely mediating protein quality control and function. Here, we conducted an integrated multiomic analysis to identify critical ubiquitination-associated molecules in intestinal I/R and identified endoplasmic reticulum-located HRD1 as a candidate molecule. During intestinal I/R, excessive ER stress plays a central role by causing apoptotic pathway activation. In particular, we found that ER stress-mediated apoptosis was mitigated by HRD1 knockdown in intestinal I/R mice. Mechanistically, TMEM2 was identified as a new substrate of HRD1 in intestinal I/R by mass spectrometry analysis, which has a crucial role in attenuating apoptosis and promoting non-canonical ER stress resistance. A strong negative correlation was found between the protein levels of HRD1 and TMEM2 in human intestinal ischemia samples. Specifically, HRD1 interacted with the lysine 42 residue of TMEM2 and reduced its stabilization by K48-linked polyubiquitination. Furthermore, KEGG pathway analysis revealed that TMEM2 regulated ER stress-mediated apoptosis in association with the PI3k/Akt signaling pathway rather than canonical ER stress pathways. In summary, HRD1 regulates ER stress-mediated apoptosis through a non-canonical pathway by ubiquitinating TMEM2 and inhibiting PI3k/Akt activation during intestinal I/R. The current study shows that HRD1 is an intestinal I/R critical regulator and that targeting the HRD1/TMEM2 axis may be a promising therapeutic approach.
Collapse
Affiliation(s)
- Xuzi Zhao
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Deshun Liu
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Zhao Chen
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Shili Ning
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China
| | - Lu Meng
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, 116044, Dalian, China.
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 116023, Dalian, China.
| |
Collapse
|
44
|
Lin LL, Wang HH, Pederson B, Wei X, Torres M, Lu Y, Li ZJ, Liu X, Mao H, Wang H, Zhou LE, Zhao Z, Sun S, Qi L. SEL1L-HRD1 interaction is required to form a functional HRD1 ERAD complex. Nat Commun 2024; 15:1440. [PMID: 38365914 PMCID: PMC10873344 DOI: 10.1038/s41467-024-45633-0] [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: 06/22/2023] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
The SEL1L-HRD1 protein complex represents the most conserved branch of endoplasmic reticulum (ER)-associated degradation (ERAD). Despite recent advances in both mouse models and humans, in vivo evidence for the importance of SEL1L in the ERAD complex formation and its (patho-)physiological relevance in mammals remains limited. Here we report that SEL1L variant p.Ser658Pro (SEL1LS658P) is a pathogenic hypomorphic mutation, causing partial embryonic lethality, developmental delay, and early-onset cerebellar ataxia in homozygous mice carrying the bi-allelic variant. Biochemical analyses reveal that SEL1LS658P variant not only reduces the protein stability of SEL1L, but attenuates the SEL1L-HRD1 interaction, likely via electrostatic repulsion between SEL1L F668 and HRD1 Y30 residues. Proteomic screens of SEL1L and HRD1 interactomes reveal that SEL1L-HRD1 interaction is a prerequisite for the formation of a functional HRD1 ERAD complex, as SEL1L is required for the recruitment of E2 enzyme UBE2J1 as well as DERLIN to HRD1. These data not only establish the disease relevance of SEL1L-HRD1 ERAD, but also provide additional insight into the formation of a functional HRD1 ERAD complex.
Collapse
Affiliation(s)
- Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Huilun Helen Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Brent Pederson
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Mauricio Torres
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - You Lu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Xiaodan Liu
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Hancheng Mao
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Hui Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Linyao Elina Zhou
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Shengyi Sun
- Department of Pharmacology, University of Virginia, School of Medicine, Charlottesville, VA, 22908, USA.
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
45
|
Wu Z, Xiao C, Long J, Huang W, You F, Li X. Mitochondrial dynamics and colorectal cancer biology: mechanisms and potential targets. Cell Commun Signal 2024; 22:91. [PMID: 38302953 PMCID: PMC10835948 DOI: 10.1186/s12964-024-01490-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal cancer (CRC) is a significant public health concern, and its development is associated with mitochondrial dysfunction. Mitochondria can adapt to the high metabolic demands of cancer cells owing to their plasticity and dynamic nature. The fusion-fission dynamics of mitochondria play a crucial role in signal transduction and metabolic functions of CRC cells. Enhanced mitochondrial fission promotes the metabolic reprogramming of CRC cells, leading to cell proliferation, metastasis, and chemoresistance. Excessive fission can also trigger mitochondria-mediated apoptosis. In contrast, excessive mitochondrial fusion leads to adenosine triphosphate (ATP) overproduction and abnormal tumor proliferation, whereas moderate fusion protects intestinal epithelial cells from oxidative stress-induced mitochondrial damage, thus preventing colitis-associated cancer (CAC). Therefore, an imbalance in mitochondrial dynamics can either promote or inhibit CRC progression. This review provides an overview of the mechanism underlying mitochondrial fusion-fission dynamics and their impact on CRC biology. This revealed the dual role of mitochondrial fusion-fission dynamics in CRC development and identified potential drug targets. Additionally, this study partially explored mitochondrial dynamics in immune and vascular endothelial cells in the tumor microenvironment, suggesting promising prospects for targeting key fusion/fission effector proteins against CRC.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing Long
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Institute of Oncology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
46
|
Wang L, Wang M, Niu H, Zhi Y, Li S, He X, Ren Z, Wen S, Wu L, Wen S, Zhang R, Wen Z, Yang J, Zhang X, Chen Y, Qian X, Shi G. Cholesterol-induced HRD1 reduction accelerates vascular smooth muscle cell senescence via stimulation of endoplasmic reticulum stress-induced reactive oxygen species. J Mol Cell Cardiol 2024; 187:51-64. [PMID: 38171043 DOI: 10.1016/j.yjmcc.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/28/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Senescence of vascular smooth muscle cells (VSMCs) is a key contributor to plaque vulnerability in atherosclerosis (AS), which is affected by endoplasmic reticulum (ER) stress and reactive oxygen species (ROS) production. However, the crosstalk between ER stress and ROS production in the pathogenesis of VSMC senescence remains to be elucidated. ER-associated degradation (ERAD) is a complex process that clears unfolded or misfolded proteins to maintain ER homeostasis. HRD1 is the major E3 ligase in mammalian ERAD machineries that catalyzes ubiquitin conjugation to the unfolded or misfolded proteins for degradation. Our results showed that HRD1 protein levels were reduced in human AS plaques and aortic roots from ApoE-/- mice fed with high-fat diet (HFD), along with the increased ER stress response. Exposure to cholesterol in VSMCs activated inflammatory signaling and induced senescence, while reduced HRD1 protein expression. CRISPR Cas9-mediated HRD1 knockout (KO) exacerbated cholesterol- and thapsigargin-induced cell senescence. Inhibiting ER stress with 4-PBA (4-Phenylbutyric acid) partially reversed the ROS production and cell senescence induced by HRD1 deficiency in VSMCs, suggesting that ER stress alone could be sufficient to induce ROS production and senescence in VSMCs. Besides, HRD1 deficiency led to mitochondrial dysfunction, and reducing ROS production from impaired mitochondria partly reversed HRD1 deficiency-induced cell senescence. Finally, we showed that the overexpression of HDR1 reversed cholesterol-induced ER stress, ROS production, and cellular senescence in VSMCs. Our findings indicate that HRD1 protects against senescence by maintaining ER homeostasis and mitochondrial functionality. Thus, targeting HRD1 function may help to mitigate VSMC senescence and prevent vascular aging related diseases. TRIAL REGISTRATION: A real-world study based on the discussion of primary and secondary prevention strategies for coronary heart disease, URL:https://www.clinicaltrials.gov, the trial registration number is [2022]-02-121-01.
Collapse
Affiliation(s)
- Linli Wang
- Department of Cardiology, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Min Wang
- Department of Cardiology, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Haiming Niu
- Department of Critical Care Medicine, Zhongshan People's Hospital, Zhongshan, Guangdong, China.
| | - Yaping Zhi
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Shasha Li
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xuemin He
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zhitao Ren
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Shiyi Wen
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Lin Wu
- Department of Cardiology, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Siying Wen
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Rui Zhang
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zheyao Wen
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Jing Yang
- Department of Endocrinology and Metabolism, The Eighth affiliated hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Ximei Zhang
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Yanming Chen
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Xiaoxian Qian
- Department of Cardiology, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Guojun Shi
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, Third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
47
|
Liu L, Wu J, Yan Y, Cheng S, Yu S, Wang Y. DERL2 (derlin 2) stabilizes BAG6 (BAG cochaperone 6) in chemotherapy resistance of cholangiocarcinoma. J Physiol Biochem 2024; 80:81-97. [PMID: 37815698 PMCID: PMC10810035 DOI: 10.1007/s13105-023-00986-w] [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: 04/01/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
DERL2 (derlin 2) is a critical component of the endoplasmic reticulum quality control pathway system whose mutations play an important role in carcinogenesis, including cholangiocarcinoma (CHOL). However, its role and its underlying mechanism have yet to be elucidated. Herein, we revealed that DERL2 was highly expressed in CHOL and considered as an independent prognostic indicator for inferior survival in CHOL. DERL2 ectopically expressed in CHOL cells promoted cell proliferation and colony formation rates, and depleting DERL2 in CHOL cells curbed tumor growth in vitro and in vivo. More interestingly, the knockout of DERL2 augmented the growth-inhibitory effect of gemcitabine chemotherapy on CHOL cells by inducing cell apoptosis. Mechanistically, we discovered that DERL2 interacted with BAG6 (BAG cochaperone 6), thereby extending its half-life and reinforcing the oncogenic role of BAG6 in CHOL progression.
Collapse
Affiliation(s)
- Luzheng Liu
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Hainan, 570311, China
| | - Jincai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, 570311, China
| | - Yanggang Yan
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Hainan, 570311, China
| | - Shoucai Cheng
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Hainan, 570311, China
| | - Shuyong Yu
- Department of Gastrointestinal Surgery, Hainan Cancer Hospital, Hainan, 570312, China.
| | - Yong Wang
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Hainan, 570311, China.
| |
Collapse
|
48
|
Wang M, Sun Z, Ou Y, Ge W, Yuan M, Xu B. Electroacupuncture Mediates Fat Metabolism and Autophagy via a Sirt3-Dependent Mechanism in Mice Fed High-Fat Diet. Adv Biol (Weinh) 2024; 8:e2300370. [PMID: 37840428 DOI: 10.1002/adbi.202300370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/02/2023] [Indexed: 10/17/2023]
Abstract
This study investigates the therapeutic potential of electroacupuncture (EA) on obesity, focusing on its influence on autophagy and energy metabolism, utilizing a high-fat diet (HFD)-induced mouse model. Treatment with EA significantly reduces body weight, fat deposition, and lipid accumulation in HFD-fed mice. Additionally, EA effectively ameliorates metabolic imbalances, reducing blood glucose levels and plasma markers of liver function. At the molecular level, EA enhances the expression of thermogenesis-associated genes in brown adipose tissue and decreases p53 expression, suggesting a decrease in apoptosis. Autophagy in white adipose tissue is inhibited by EA, as demonstrated by the suppression of key autophagy-related proteins. Further experiments highlight the critical role of Sirtuin 3 (Sirt3) in EA's anti-obesity effects. Sirt3 supplementation combined with EA results in reduced body weight, fat deposition, and lipid accumulation, along with modulations in key metabolic indicators. Moreover, EA's modulatory effect on uncoupling protein 1 (Ucp1), Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Pgc-1α), and p53 is found to be Sirt3 dependent. In conclusion, EA exerts beneficial effects against obesity through Sirt3-dependent modulation of autophagy and energy metabolism, indicating a potential therapeutic approach for obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Ming Wang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
- Department of Acupuncture and Massage, Geriatric Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210024, China
| | - Zhicheng Sun
- Department of Acupuncture and Massage, Geriatric Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210024, China
| | - Yanggang Ou
- Department of Acupuncture and Massage, Geriatric Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210024, China
| | - Wei Ge
- Department of Acupuncture and Massage, Geriatric Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210024, China
| | - Mengqian Yuan
- Department of Acupuncture Rehabilitation, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, 210024, China
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
49
|
Wei X, Lu Y, Lin LL, Zhang C, Chen X, Wang S, Wu SA, Li ZJ, Quan Y, Sun S, Qi L. Proteomic screens of SEL1L-HRD1 ER-associated degradation substrates reveal its role in glycosylphosphatidylinositol-anchored protein biogenesis. Nat Commun 2024; 15:659. [PMID: 38253565 PMCID: PMC10803770 DOI: 10.1038/s41467-024-44948-2] [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] [Received: 06/15/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) plays indispensable roles in many physiological processes; however, the nature of endogenous substrates remains largely elusive. Here we report a proteomics strategy based on the intrinsic property of the SEL1L-HRD1 ERAD complex to identify endogenous ERAD substrates both in vitro and in vivo. Following stringent filtering using a machine learning algorithm, over 100 high-confidence potential substrates are identified in human HEK293T and mouse brown adipose tissue, among which ~88% are cell type-specific. One of the top shared hits is the catalytic subunit of the glycosylphosphatidylinositol (GPI)-transamidase complex, PIGK. Indeed, SEL1L-HRD1 ERAD attenuates the biogenesis of GPI-anchored proteins by specifically targeting PIGK for proteasomal degradation. Lastly, several PIGK disease variants in inherited GPI deficiency disorders are also SEL1L-HRD1 ERAD substrates. This study provides a platform and resources for future effort to identify proteome-wide endogenous substrates in vivo, and implicates SEL1L-HRD1 ERAD in many cellular processes including the biogenesis of GPI-anchored proteins.
Collapse
Affiliation(s)
- Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - You Lu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Chengxin Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Xinxin Chen
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Siwen Wang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Shuangcheng Alivia Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Yujun Quan
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Shengyi Sun
- Department of Pharmacology, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
50
|
Umphred-Wilson K, Adoro S. Hypomorphic human SEL1L and HRD1 variants uncouple multilayered ER-associated degradation machinery. J Clin Invest 2024; 134:e175448. [PMID: 38226624 PMCID: PMC10786685 DOI: 10.1172/jci175448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
The suppressor of lin-12-like-HMG-CoA reductase degradation 1 (SEL1L-HRD1) complex of the endoplasmic reticulum-associated degradation (ERAD) machinery is a key cellular proteostasis pathway. Although previous studies have shown ERAD as promoting the development and maintenance of many cell types in mice, its importance to human physiology remained undetermined. In two articles in this issue of the JCI, Qi and colleagues describe four biallelic hypomorphic SEL1L and HRD1 variants that were associated with neurodevelopment disorders, locomotor dysfunction, impaired immunity, and premature death in patients. These pathogenic SEL1L-HRD1 variants shine a light on the critical importance of ERAD in humans and pave the way for future studies dissecting ERAD mechanisms in specific cell types.
Collapse
|