1
|
Sun Z, Cheng X, Wang Z, Qiao C, Qian H, Yuan T, Lv Z, Sun W, Zhang H, Liu Y, Lu Z, Lin J, Lai C, Wang Y, Yang X, Wang X, Meng J, Bao N. Single-nucleus transcriptomics reveals subsets of degenerative myonuclei after rotator cuff tear-induced muscle atrophy. Cell Prolif 2025; 58:e13763. [PMID: 39435630 PMCID: PMC11882757 DOI: 10.1111/cpr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Rotator cuff tear (RCT) is the primary cause of shoulder pain and disability and frequently trigger muscle degeneration characterised by muscle atrophy, fatty infiltration and fibrosis. Single-nucleus RNA sequencing (snRNA-seq) was used to reveal the transcriptional changes in the supraspinatus muscle after RCT. Supraspinatus muscles were obtained from patients with habitual shoulder dislocation (n = 3) and RCT (n = 3). In response to the RCT, trajectory analysis showed progression from normal myonuclei to ANKRD1+ myonuclei, which captured atrophy-and fatty infiltration-related regulons (KLF5, KLF10, FOSL1 and BHLHE40). Transcriptomic alterations in fibro/adipogenic progenitors (FAPs) and muscle satellite cells (MuSCs) have also been studied. By predicting cell-cell interactions, we observed communication alterations between myofibers and muscle-resident cells following RCT. Our findings reveal the plasticity of muscle cells in response to RCT and offer valuable insights into the molecular mechanisms and potential therapeutic targets of RCT.
Collapse
Affiliation(s)
- Ziying Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xi Cheng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zheng Wang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chenfeng Qiao
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Hong Qian
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhongyang Lv
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Wenshuang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Hanwen Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yuan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Zhihao Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Jintao Lin
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Chengteng Lai
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Yang Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xiaojiang Yang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
| | - Xingyun Wang
- Hongqiao International Institute of Medicine, Tongren HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Jia Meng
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| | - Nirong Bao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsuPeople's Republic of China
- Department of Orthopedics, Jinling Clinical Medical CollegeNanjing University of Chinese MedicineNanjingJiangsuPeople's Republic of China
| |
Collapse
|
2
|
Tomaz da Silva M, Joshi AS, Kumar A. Fibroblast growth factor-inducible 14 regulates satellite cell self-renewal and expansion during skeletal muscle repair. JCI Insight 2025; 10:e187825. [PMID: 39874107 PMCID: PMC11949035 DOI: 10.1172/jci.insight.187825] [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: 10/08/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Skeletal muscle regeneration in adults is predominantly driven by satellite cells. Loss of satellite cell pool and function leads to skeletal muscle wasting in many conditions and disease states. Here, we demonstrate that the levels of fibroblast growth factor-inducible 14 (Fn14) were increased in satellite cells after muscle injury. Conditional ablation of Fn14 in Pax7-expressing satellite cells drastically reduced their expansion and skeletal muscle regeneration following injury. Fn14 was required for satellite cell self-renewal and proliferation as well as to prevent precocious differentiation. Targeted deletion of Fn14 inhibited Notch signaling but led to the spurious activation of STAT3 signaling in regenerating skeletal muscle and in cultured muscle progenitor cells. Silencing of STAT3 improved proliferation and inhibited premature differentiation of Fn14-deficient satellite cells. Furthermore, conditional ablation of Fn14 in satellite cells exacerbated myopathy in the mdx mouse model of Duchenne muscular dystrophy (DMD), whereas its overexpression improved the engraftment of exogenous muscle progenitor cells into the dystrophic muscle of mdx mice. Altogether, our study highlights the crucial role of Fn14 in the regulation of satellite cell fate and function and suggests that Fn14 can be a potential molecular target to improve muscle regeneration in muscular disorders.
Collapse
MESH Headings
- Animals
- Satellite Cells, Skeletal Muscle/metabolism
- Mice
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/injuries
- Muscle, Skeletal/physiology
- Regeneration/physiology
- Mice, Inbred mdx
- Cell Differentiation
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- TWEAK Receptor/metabolism
- TWEAK Receptor/genetics
- Cell Proliferation
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Signal Transduction
- Cell Self Renewal
- Disease Models, Animal
- PAX7 Transcription Factor/metabolism
- Male
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Meiricris Tomaz da Silva
- Institute of Muscle Biology and Cachexia, and
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Aniket S. Joshi
- Institute of Muscle Biology and Cachexia, and
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Ashok Kumar
- Institute of Muscle Biology and Cachexia, and
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas, USA
| |
Collapse
|
3
|
Tomaz da Silva M, Joshi AS, Kumar A. Fibroblast growth factor-inducible 14 regulates satellite cell self-renewal and expansion during skeletal muscle repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.06.616900. [PMID: 39803454 PMCID: PMC11722277 DOI: 10.1101/2024.10.06.616900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Skeletal muscle regeneration in adults is predominantly driven by satellite cells. Loss of satellite cell pool and function leads to skeletal muscle wasting in many conditions and disease states. Here, we demonstrate that the levels of fibroblast growth factor-inducible 14 (Fn14) are increased in satellite cells after muscle injury. Conditional ablation of Fn14 in Pax7-expressing satellite cells drastically reduces their expansion and skeletal muscle regeneration following injury. Fn14 is required for satellite cell self-renewal and proliferation as well as to prevent precocious differentiation. Targeted deletion of Fn14 inhibits Notch signaling but leads to the spurious activation of STAT3 signaling in regenerating skeletal muscle and in cultured muscle progenitor cells. Silencing of STAT3 improves proliferation and inhibits premature differentiation of Fn14-deficient satellite cells. Furthermore, conditional ablation of Fn14 in satellite cells exacerbates myopathy in the mdx mouse model of Duchenne muscular dystrophy (DMD) whereas its overexpression improves the engraftment of exogenous muscle progenitor cells into the dystrophic muscle of mdx mice. Altogether, our study highlights the crucial role of Fn14 in the regulation of satellite cell fate and function and suggests that Fn14 can be a potential molecular target to improve muscle regeneration in muscular disorders.
Collapse
Affiliation(s)
- Meiricris Tomaz da Silva
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Aniket S. Joshi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Ashok Kumar
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| |
Collapse
|
4
|
Joshi AS, Castillo MB, Tomaz da Silva M, Vuong AT, Gunaratne PH, Darabi R, Liu Y, Kumar A. Single-nucleus transcriptomic analysis reveals the regulatory circuitry of myofiber XBP1 during regenerative myogenesis. iScience 2024; 27:111372. [PMID: 39650729 PMCID: PMC11625362 DOI: 10.1016/j.isci.2024.111372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/11/2024] Open
Abstract
Endoplasmic reticulum (ER) stress-induced unfolded protein response (UPR) is activated in skeletal muscle under multiple conditions. However, the role of the UPR in the regulation of muscle regeneration remains less understood. We demonstrate that gene expression of various markers of the UPR is induced in both myogenic and non-myogenic cells in regenerating muscle. Genetic ablation of X-box binding protein 1 (XBP1), a downstream target of the Inositol requiring enzyme 1α (IRE1α) arm of the UPR, in myofibers attenuates muscle regeneration in adult mice. Single nucleus RNA sequencing (snRNA-seq) analysis showed that deletion of XBP1 in myofibers perturbs proteolytic systems and mitochondrial function in myogenic cells. Trajectory analysis of snRNA-seq dataset showed that XBP1 regulates the abundance of satellite cells and the formation of new myofibers in regenerating muscle. In addition, ablation of XBP1 disrupts the composition of non-myogenic cells in injured muscle microenvironment. Collectively, our study suggests that myofiber XBP1 regulates muscle regeneration through both cell-autonomous and -non-autonomous mechanisms.
Collapse
Affiliation(s)
- Aniket S. Joshi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Micah B. Castillo
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Meiricris Tomaz da Silva
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Anh Tuan Vuong
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Preethi H. Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Radbod Darabi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Yu Liu
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Ashok Kumar
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX 77204, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| |
Collapse
|
5
|
Chen K, Shoulders MD. Protein Glycosylation Patterns Shaped By the IRE1-XBP1s Arm of the Unfolded Protein Response. Isr J Chem 2024; 64:e202300162. [PMID: 40083477 PMCID: PMC11906193 DOI: 10.1002/ijch.202300162] [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: 11/15/2023] [Indexed: 03/16/2025]
Abstract
The unfolded protein response (UPR) is a sensing and signaling pathway that surveys the endoplasmic reticulum (ER) for protein folding challenges and responds whenever issues are detected. UPR activation leads to upregulation of secretory pathway chaperones and quality control factors, as well as reduces the nascent protein load on the ER, thereby restoring and maintaining proteostasis. This paradigm-defining view of the role of the UPR is accurate, but it elides additional key functions of the UPR in cell biology. In particular, recent work has revealed that the UPR can shape the structure and function of N- and O-glycans installed on ER client proteins. This crosstalk between the UPR's response to protein misfolding and the regulation of glycosylation remains insufficiently understood. Still, emerging evidence makes it clear that the UPR, and particularly the IRE1-XBP1s arm of the UPR, may be a central regulator of protein glycosylation with important biological consequences. In this review, we discuss the crosstalk between proteostasis, the UPR, and glycosylation, present progress towards understanding biological functions of this crosstalk, and examine potential roles in diseases such as cancer.
Collapse
Affiliation(s)
- Kenny Chen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
Takasugi M, Nonaka Y, Takemura K, Yoshida Y, Stein F, Schwarz JJ, Adachi J, Satoh J, Ito S, Tombline G, Biashad SA, Seluanov A, Gorbunova V, Ohtani N. An atlas of the aging mouse proteome reveals the features of age-related post-transcriptional dysregulation. Nat Commun 2024; 15:8520. [PMID: 39353907 PMCID: PMC11445428 DOI: 10.1038/s41467-024-52845-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: 07/03/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
To what extent and how post-transcriptional dysregulation affects aging proteome remains unclear. Here, we provide proteomic data of whole-tissue lysates (WTL) and low-solubility protein-enriched fractions (LSF) of major tissues collected from mice of 6, 15, 24, and 30 months of age. Low-solubility proteins are preferentially affected by age and the analysis of LSF doubles the number of proteins identified to be differentially expressed with age. Simultaneous analysis of proteome and transcriptome using the same tissue homogenates reveals the features of age-related post-transcriptional dysregulation. Post-transcriptional dysregulation becomes evident especially after 24 months of age and age-related post-transcriptional dysregulation leads to accumulation of core matrisome proteins and reduction of mitochondrial membrane proteins in multiple tissues. Based on our in-depth proteomic data and sample-matched transcriptome data of adult, middle-aged, old, and geriatric mice, we construct the Mouse aging proteomic atlas ( https://aging-proteomics.info/ ), which provides a thorough and integrative view of age-related gene expression changes.
Collapse
Affiliation(s)
- Masaki Takasugi
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan.
| | - Yoshiki Nonaka
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Kazuaki Takemura
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Yuya Yoshida
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan
| | - Frank Stein
- Proteomic Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | | | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Junko Satoh
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Gregory Tombline
- Department of Biology, University of Rochester, Rochester, NY, USA
| | | | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Naoko Ohtani
- Department of Pathophysiology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
7
|
Joshi AS, Tomaz da Silva M, Roy A, Koike TE, Wu M, Castillo MB, Gunaratne PH, Liu Y, Iwawaki T, Kumar A. The IRE1α/XBP1 signaling axis drives myoblast fusion in adult skeletal muscle. EMBO Rep 2024; 25:3627-3650. [PMID: 38982191 PMCID: PMC11316051 DOI: 10.1038/s44319-024-00197-4] [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: 11/22/2023] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024] Open
Abstract
Skeletal muscle regeneration involves a signaling network that regulates the proliferation, differentiation, and fusion of muscle precursor cells to injured myofibers. IRE1α, one of the arms of the unfolded protein response, regulates cellular proteostasis in response to ER stress. Here, we demonstrate that inducible deletion of IRE1α in satellite cells of mice impairs skeletal muscle regeneration through inhibiting myoblast fusion. Knockdown of IRE1α or its downstream target, X-box protein 1 (XBP1), also inhibits myoblast fusion during myogenesis. Transcriptome analysis revealed that knockdown of IRE1α or XBP1 dysregulates the gene expression of molecules involved in myoblast fusion. The IRE1α-XBP1 axis mediates the gene expression of multiple profusion molecules, including myomaker (Mymk). Spliced XBP1 (sXBP1) transcription factor binds to the promoter of Mymk gene during myogenesis. Overexpression of myomaker in IRE1α-knockdown cultures rescues fusion defects. Inducible deletion of IRE1α in satellite cells also inhibits myoblast fusion and myofiber hypertrophy in response to functional overload. Collectively, our study demonstrates that IRE1α promotes myoblast fusion through sXBP1-mediated up-regulation of the gene expression of multiple profusion molecules, including myomaker.
Collapse
Affiliation(s)
- Aniket S Joshi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Meiricris Tomaz da Silva
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Anirban Roy
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Tatiana E Koike
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Mingfu Wu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Micah B Castillo
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA.
| |
Collapse
|
8
|
Tomaz da Silva M, Joshi AS, Castillo MB, Koike TE, Roy A, Gunaratne PH, Kumar A. Fn14 promotes myoblast fusion during regenerative myogenesis. Life Sci Alliance 2023; 6:e202302312. [PMID: 37813488 PMCID: PMC10561765 DOI: 10.26508/lsa.202302312] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Skeletal muscle regeneration involves coordinated activation of an array of signaling pathways. Fibroblast growth factor-inducible 14 (Fn14) is a bona fide receptor for the TWEAK cytokine. Levels of Fn14 are increased in the skeletal muscle of mice after injury. However, the cell-autonomous role of Fn14 in muscle regeneration remains unknown. Here, we demonstrate that global deletion of the Fn14 receptor in mice attenuates muscle regeneration. Conditional ablation of Fn14 in myoblasts but not in differentiated myofibers of mice inhibits skeletal muscle regeneration. Fn14 promotes myoblast fusion without affecting the levels of myogenic regulatory factors in the regenerating muscle. Fn14 deletion in myoblasts hastens initial differentiation but impairs their fusion. The overexpression of Fn14 in myoblasts results in the formation of myotubes having an increased diameter after induction of differentiation. Ablation of Fn14 also reduces the levels of various components of canonical Wnt and calcium signaling both in vitro and in vivo. Forced activation of Wnt signaling rescues fusion defects in Fn14-deficient myoblast cultures. Collectively, our results demonstrate that Fn14-mediated signaling positively regulates myoblast fusion and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Meiricris Tomaz da Silva
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Aniket S Joshi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Micah B Castillo
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Tatiana E Koike
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Anirban Roy
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| |
Collapse
|
9
|
Liang L, Zhang F, Feng N, Kuang B, Fan M, Chen C, Pan Y, Zhou P, Geng N, Li X, Xian M, Deng L, Li X, Kuang L, Luo F, Tan Q, Xie Y, Guo F. IRE1α protects against osteoarthritis by regulating progranulin-dependent XBP1 splicing and collagen homeostasis. Exp Mol Med 2023; 55:2376-2389. [PMID: 37907740 PMCID: PMC10689778 DOI: 10.1038/s12276-023-01106-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: 12/20/2022] [Revised: 07/17/2023] [Accepted: 08/02/2023] [Indexed: 11/02/2023] Open
Abstract
Osteoarthritis (OA) is a full-joint, multifactorial, degenerative and inflammatory disease that seriously affects the quality of life of patients due to its disabling and pain-causing properties. ER stress has been reported to be closely related to the progression of OA. The inositol-requiring enzyme 1α/X-box-binding protein-1 spliced (IRE1α/XBP1s) pathway, which is highly expressed in the chondrocytes of OA patients, promotes the degradation and refolding of abnormal proteins during ER stress and maintains the stability of the ER environment of chondrocytes, but its function and the underlying mechanisms of how it contributes to the progression of OA remain unclear. This study investigates the role of IRE1α/ERN1 in OA. Specific deficiency of ERN1 in chondrocytes spontaneously resulted in OA-like cartilage destruction and accelerated OA progression in a surgically induced arthritis model. Local delivery of AdERN1 relieved degradation of the cartilage matrix and prevented OA development in an ACLT-mediated model. Mechanistically, progranulin (PGRN), an intracellular chaperone, binds to IRE1α, promoting its phosphorylation and splicing of XBP1u to generate XBP1s. XBP1s protects articular cartilage through TNF-α/ERK1/2 signaling and further maintains collagen homeostasis by regulating type II collagen expression. The chondroprotective effect of IRE1α/ERN1 is dependent on PGRN and XBP1s splicing. ERN1 deficiency accelerated cartilage degeneration in OA by reducing PGRN expression and XBP1s splicing, subsequently decreasing collagen II expression and triggering collagen structural abnormalities and an imbalance in collagen homeostasis. This study provides new insights into OA pathogenesis and the UPR and suggests that IRE1α/ERN1 may serve as a potential target for the treatment of joint degenerative diseases, including OA.
Collapse
Affiliation(s)
- Li Liang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Fengmei Zhang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
- Laboratory Animal Center, Chongqing Medical University, 400016, Chongqing, China
| | - Naibo Feng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Biao Kuang
- Department of Orthopedics, The 2nd Affiliated Hospital of Chongqing Medical University, 400072, Chongqing, China
| | - Mengtian Fan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Cheng Chen
- Department of Orthopedics, The 1st Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Yiming Pan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Pengfei Zhou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, College of Stomatology, Chongqing Medical University, 400016, Chongqing, China
| | - Nana Geng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Xingyue Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Menglin Xian
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Lin Deng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoli Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Liang Kuang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Fengjin Guo
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
10
|
Abdon B, Liang Y, da Luz Scheffer D, Torres M, Shrestha N, Reinert RB, Lu Y, Pederson B, Bugarin-Lapuz A, Kersten S, Qi L. Muscle-specific ER-associated degradation maintains postnatal muscle hypertrophy and systemic energy metabolism. JCI Insight 2023; 8:e170387. [PMID: 37535424 PMCID: PMC10578429 DOI: 10.1172/jci.insight.170387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023] Open
Abstract
The growth of skeletal muscle relies on a delicate equilibrium between protein synthesis and degradation; however, how proteostasis is managed in the endoplasmic reticulum (ER) is largely unknown. Here, we report that the SEL1L-HRD1 ER-associated degradation (ERAD) complex, the primary molecular machinery that degrades misfolded proteins in the ER, is vital to maintain postnatal muscle growth and systemic energy balance. Myocyte-specific SEL1L deletion blunts the hypertrophic phase of muscle growth, resulting in a net zero gain of muscle mass during this developmental period and a 30% reduction in overall body growth. In addition, myocyte-specific SEL1L deletion triggered a systemic reprogramming of metabolism characterized by improved glucose sensitivity, enhanced beigeing of adipocytes, and resistance to diet-induced obesity. These effects were partially mediated by the upregulation of the myokine FGF21. These findings highlight the pivotal role of SEL1L-HRD1 ERAD activity in skeletal myocytes for postnatal muscle growth, and its physiological integration in maintaining whole-body energy balance.
Collapse
Affiliation(s)
- Benedict Abdon
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yusheng Liang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Débora da Luz Scheffer
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Mauricio Torres
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Neha Shrestha
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rachel B. Reinert
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - You Lu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brent Pederson
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Amara Bugarin-Lapuz
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sander Kersten
- Nutrition Metabolism and Genomics group, Wageningen University, Wageningen, Netherlands
| | - Ling Qi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Martinez-Amaro FJ, Garcia-Padilla C, Franco D, Daimi H. LncRNAs and CircRNAs in Endoplasmic Reticulum Stress: A Promising Target for Cardiovascular Disease? Int J Mol Sci 2023; 24:9888. [PMID: 37373035 DOI: 10.3390/ijms24129888] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The endoplasmic reticulum (ER) is a principal subcellular organelle responsible for protein quality control in the secretory pathway, preventing protein misfolding and aggregation. Failure of protein quality control in the ER triggers several molecular mechanisms such as ER-associated degradation (ERAD), the unfolded protein response (UPR) or reticulophagy, which are activated upon ER stress (ERS) to re-establish protein homeostasis by transcriptionally and translationally regulated complex signalling pathways. However, maintenance over time of ERS leads to apoptosis if such stress cannot be alleviated. The presence of abnormal protein aggregates results in loss of cardiomyocyte protein homeostasis, which in turn results in several cardiovascular diseases such as dilated cardiomyopathy (DCM) or myocardial infarction (MI). The influence of a non-coding genome in the maintenance of proper cardiomyocyte homeostasis has been widely proven. To date, the impact of microRNAs in molecular mechanisms orchestrating ER stress response has been widely described. However, the role of long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) is just beginning to be addressed given the potential role of these RNA classes as therapeutic molecules. Here, we provide a current state-of-the-art review of the roles of distinct lncRNAs and circRNAs in the modulation of ERS and UPR and their impact in cardiovascular diseases.
Collapse
Affiliation(s)
| | - Carlos Garcia-Padilla
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Houria Daimi
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
- Department of Biology, Faculty of Sciences, University of Gabes, Gabes 6072, Tunisia
| |
Collapse
|
12
|
Sousa NS, Brás MF, Antunes IB, Lindholm P, Neves J, Sousa-Victor P. Aging disrupts MANF-mediated immune modulation during skeletal muscle regeneration. NATURE AGING 2023; 3:585-599. [PMID: 37118549 DOI: 10.1038/s43587-023-00382-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/16/2023] [Indexed: 04/30/2023]
Abstract
Age-related decline in skeletal muscle regenerative capacity is multifactorial, yet the contribution of immune dysfunction to regenerative failure is unknown. Macrophages are essential for effective debris clearance and muscle stem cell activity during muscle regeneration, but the regulatory mechanisms governing macrophage function during muscle repair are largely unexplored. Here, we uncover a new mechanism of immune modulation operating during skeletal muscle regeneration that is disrupted in aged animals and relies on the regulation of macrophage function. The immune modulator mesencephalic astrocyte-derived neurotrophic factor (MANF) is induced following muscle injury in young mice but not in aged animals, and its expression is essential for regenerative success. Regenerative impairments in aged muscle are associated with defects in the repair-associated myeloid response similar to those found in MANF-deficient models and could be improved through MANF delivery. We propose that restoring MANF levels is a viable strategy to improve myeloid response and regenerative capacity in aged muscle.
Collapse
Affiliation(s)
- Neuza S Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Margarida F Brás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Inês B Antunes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Päivi Lindholm
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Joana Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Sousa-Victor
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
13
|
Wang B, Zhou W, Zhang H, Wang W, Zhang B, Li S. Exploring the effect of Weifuchun capsule on the toll-like receptor pathway mediated HES6 and immune regulation against chronic atrophic gastritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115930. [PMID: 36403744 DOI: 10.1016/j.jep.2022.115930] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Weifuchun capsule (WFC) is a traditional Chinese patent medicine for chronic atrophic gastritis (CAG) in clinic. However, the mechanism of action of WFC for CAG still remains unclear due to its complex composition. AIM OF THE STUDY The study was projected to uncover the mechanism of action of WFC and the corresponding pharmacodynamic substance of WFC against CAG as well as providing a standard example for the research of traditional Chinese medicine (TCM) from the perspective of the network and the system. MATERIALS AND METHODS We identified the compounds of WFC through LC-MS/MS analysis and performed a systematic network targets analysis for WFC in the treatment of CAG which thoroughly described the mechanism of action of WFC for CAG. Based on analysis integrating omics data and algorithms, we focused on the specific immune regulatory role of WFC in the treatment of CAG, especially on a hub pathway, Toll-like receptor signaling pathway and thus deciphered the role of WFC in immune regulation, anti-inflammation and mediation of HES6. In experiments part, MNNG-GES-1-cell line and rat models were used to validate our findings. RESULTS In this study, compounds of WFC are identified through LC‒MS/MS and network target analysis is performed to dissect the specific immunoregulatory effect as well as mediation of HES6, a newly discovered biomolecule related to gastritis carcinoma progression, of WFC on CAG through the Toll-like receptor signaling pathway. Based on cell line and rat models, we verify the mechanism of action of WFC for CAG in inhibiting inflammatory cytokines, regulating immune cells like T cells and macrophages, related genes including TLR2 and CD14. It is also validated that WFC inhibits the expression of HES6 (P < 0.05). CONCLUSION Based on the combination of computational strategy and experiments, our study offers a comprehensive analysis to reveal the role of WFC in regulating immune response, inhibiting inflammation in the treatment of CAG, and provides a standard example for the research of TCM from the perspective of the network and the system.
Collapse
Affiliation(s)
- Boyang Wang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, 100084, Beijing, China
| | - Wuai Zhou
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, 100084, Beijing, China
| | - Huan Zhang
- TCM Network Pharmacology Department, Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, 300457, Tianjin, China
| | - Weihua Wang
- Center of Pharmaceutical Technology, Tsinghua University, China
| | - Bo Zhang
- TCM Network Pharmacology Department, Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, 300457, Tianjin, China.
| | - Shao Li
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
14
|
Li Z, Xu T, Peng L, Tang X, Chi Q, Li M, Li S. Polystyrene nanoplastics aggravates lipopolysaccharide-induced apoptosis in mouse kidney cells by regulating IRE1/XBP1 endoplasmic reticulum stress pathway via oxidative stress. J Cell Physiol 2023; 238:151-164. [PMID: 36370432 DOI: 10.1002/jcp.30913] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/03/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022]
Abstract
Nanoplastics (NPs) pollution poses a huge threat to the ecosystem and has become one of the environmental pollutants that have attracted much attention. There is increasing evidence that both oxidative stress and endoplasmic reticulum stress (ERS) are associated with polystyrene nanoplastics (PS-NPs) exposure. Lipopolysaccharide (LPS) has been shown to induce apoptotic damage in various tissues, but whether PS-NPs can aggravate LPS-induced apoptosis in mouse kidneys through oxidative stress-regulated inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) ERS pathway remains unclear. In this study, based on the establishment of in vitro and in vivo PS-NPs and LPS exposure models alone and in combination in mice and HEK293 cells, the effects and mechanisms of PS-NPs on LPS-induced renal cell apoptosis were investigated. The results showed that PS-NPs could aggravate LPS-induced apoptosis. PS-NPs/LPS can induce ERS through oxidative stress, activate the IRE1/XBP1 pathway, and promote the expression of apoptosis markers (Caspase-3 and Caspase-12). Kidney oxidative stress, ERS, and apoptosis in PS-NPs + LPS combined exposure group were more severe than those in the single exposure group. Interestingly, 4-phenylbutyric acid-treated HEK293 cells inhibited the expression of the IRE1/XBP1 ERS pathway and apoptotic factors in the PS-NPs + LPS combined exposure group. N-acetyl-L-cysteine effectively blocked the activation of the IRE1/XBP1 ERS pathway, suggesting that PS-NPs-induced oxidative stress is an early event that triggers ERS. Collectively, these results confirmed that PS-NPs aggravated LPS-induced apoptosis through the oxidative stress-induced IRE1/XBP1 ERS pathway. Our study provides new insights into the health threats of PS-NPs exposed to mammals and humans.
Collapse
Affiliation(s)
- Zhe Li
- Department of Animal Physiology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P.R. China
| | - Tong Xu
- Department of Animal Physiology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P.R. China
| | - Lin Peng
- Department of Animal Physiology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P.R. China
| | - Xinyu Tang
- Department of Animal Physiology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P.R. China
| | - Qianru Chi
- Department of Animal Physiology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P.R. China
| | - Ming Li
- Department of Animal Ecology, College of Life and environmental Science, Wenzhou University, Wenzhou, P.R. China
| | - Shu Li
- Department of Animal Physiology, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P.R. China
| |
Collapse
|
15
|
del Rio Oliva M, Basler M. Valosin-containing protein (VCP/p97) inhibition reduces viral clearance and induces toxicity associated with muscular damage. Cell Death Dis 2022; 13:1015. [PMID: 36456548 PMCID: PMC9715549 DOI: 10.1038/s41419-022-05461-w] [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: 08/26/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
Valosin-containing protein (VCP)/p97 has emerged as a central regulator of the ubiquitin-proteasome system by connecting ubiquitylation and degradation. The development of CB-5083, an ATPase D2-domain-selective and orally bioavailable inhibitor of VCP/p97, allows targeting of the ubiquitin-proteasome system in human diseases. In this study, we evaluated the effect of CB-5083 on the immune response in mice by using the lymphocytic choriomeningitis virus (LCMV) as an infection model. We demonstrate that LCMV infection increased the susceptibility to CB-5083 treatment in a CD8-independent manner. Administration of CB-5083 to mice reduced the cytotoxic T cell response and impaired viral clearance. Compared to uninfected cells, CB-5083 treatment enhanced the unfolded protein response in LCMV-infected cells. Administration of CB-5083 during the expansion of CD8+ T cells led to strong toxicity in mice within hours, which resulted in enhanced IL-6 levels in the serum and accumulation of poly-ubiquitinated proteins. Furthermore, we linked the observed toxicity to the specific formation of aggregates in the skeletal muscle tissue and the upregulation of both lactate dehydrogenase and creatine kinase in the serum.
Collapse
Affiliation(s)
- Marta del Rio Oliva
- grid.9811.10000 0001 0658 7699Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Michael Basler
- grid.9811.10000 0001 0658 7699Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany ,grid.469411.fBiotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| |
Collapse
|
16
|
Resistance Training Modulates Reticulum Endoplasmic Stress, Independent of Oxidative and Inflammatory Responses, in Elderly People. Antioxidants (Basel) 2022; 11:antiox11112242. [DOI: 10.3390/antiox11112242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is related to changes in the redox status, low-grade inflammation, and decreased endoplasmic reticulum unfolded protein response (UPR). Exercise has been shown to regulate the inflammatory response, balance redox homeostasis, and ameliorate the UPR. This work aimed to investigate the effects of resistance training on changes in the UPR, oxidative status, and inflammatory responses in peripheral blood mononuclear cells of elderly subjects. Thirty elderly subjects volunteered to participate in an 8-week resistance training program, and 11 youth subjects were included for basal assessments. Klotho, heat shock protein 60 (HSP60), oxidative marker expression (catalase, glutathione, lipid peroxidation, nuclear factor erythroid 2-related factor 2, protein carbonyls, reactive oxygen species, and superoxide dismutase 1 and 2), the IRE1 arm of UPR, and TLR4/TRAF6/pIRAK1 pathway activation were evaluated before and following training. No changes in the HSP60 and Klotho protein content, oxidative status markers, and TLR4/TRAF6/pIRAK1 pathway activation were found with exercise. However, an attenuation of the reduced pIRE1/IRE1 ratio was observed following training. Systems biology analysis showed that a low number of proteins (RPS27A, SYVN1, HSPA5, and XBP1) are associated with IRE1, where XBP1 and RPS27A are essential nodes according to the centrality analysis. Additionally, a gene ontology analysis confirms that endoplasmic reticulum stress is a key mechanism modulated by IRE1. These findings might partially support the modulatory effect of resistance training on the endoplasmic reticulum in the elderly.
Collapse
|