1
|
Li P, Zheng C, Hu J, Lu W, Wang D, Hao X, Zhao C, Yang L, Luo Z, Jie Q. Melatonin ameliorates Slc26a2-associated chondrodysplasias by attenuating endoplasmic reticulum stress and apoptosis of chondrocytes. Genes Dis 2025; 12:101350. [PMID: 39759111 PMCID: PMC11697235 DOI: 10.1016/j.gendis.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/11/2024] [Indexed: 01/07/2025] Open
Abstract
Although the pathogenesis and mechanism of congenital skeletal dysplasia are better understood, progress in drug development and intervention research remains limited. Here we report that melatonin treatment elicits a mitigating effect on skeletal abnormalities caused by SLC26A2 deficiency. In addition to our previous finding of endoplasmic reticulum stress upon SLC26A2 deficiency, we found calcium (Ca2+) overload jointly contributed to SLC26A2-associated chondrodysplasias. Continuous endoplasmic reticulum stress and cytosolic Ca2+ overload in turn triggered apoptosis of growth plate chondrocytes. Melatonin, known for its anti-oxidant and anti-inflammatory properties, emerged as a promising therapeutic approach in our study, which enhanced survival, proliferation, and maturation of chondrocytes by attenuating endoplasmic reticulum stress and Ca2+ overload. Our findings not only demonstrated the efficacy of melatonin in ameliorating abnormal function and cell fate of SLC26A2-deficient chondrocytes in vitro but also underscored its role in partially alleviating the skeletal dysplasia seen in Col2a1-CreER T2 ; Slc26a2 fl/fl mice. As revealed by histology and micro-CT analyses, melatonin significantly improved retarded cartilage growth, defective trabecular bone formation, and tibial genu varum in vivo. Collectively, these data shed translational insights for drug development and support melatonin as a potential treatment for SLC26A2-related chondrodysplasias.
Collapse
Affiliation(s)
- Pan Li
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Chao Zheng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jingyan Hu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Weiguang Lu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Dong Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xue Hao
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710032, China
| | - Chengxiang Zhao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhuojing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qiang Jie
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710032, China
- Research Center for Skeletal Developmental Deformity and Injury Repair, College of Life Sciences and Medicine, Northwestern University, Xi'an, Shaanxi 710032, China
- Xi'an Key Laboratory of Skeletal Development Deformity and Injury Repair, Xi'an, Shaanxi 710032, China
| |
Collapse
|
2
|
Acosta-Alvear D, Harnoss JM, Walter P, Ashkenazi A. Homeostasis control in health and disease by the unfolded protein response. Nat Rev Mol Cell Biol 2025; 26:193-212. [PMID: 39501044 DOI: 10.1038/s41580-024-00794-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 02/27/2025]
Abstract
Cells rely on the endoplasmic reticulum (ER) to fold and assemble newly synthesized transmembrane and secretory proteins - essential for cellular structure-function and for both intracellular and intercellular communication. To ensure the operative fidelity of the ER, eukaryotic cells leverage the unfolded protein response (UPR) - a stress-sensing and signalling network that maintains homeostasis by rebalancing the biosynthetic capacity of the ER according to need. The metazoan UPR can also redirect signalling from cytoprotective adaptation to programmed cell death if homeostasis restoration fails. As such, the UPR benefits multicellular organisms by preserving optimally functioning cells while removing damaged ones. Nevertheless, dysregulation of the UPR can be harmful. In this Review, we discuss the UPR and its regulatory processes as a paradigm in health and disease. We highlight important recent advances in molecular and mechanistic understanding of the UPR that enable greater precision in designing and developing innovative strategies to harness its potential for therapeutic gain. We underscore the rheostatic character of the UPR, its contextual nature and critical open questions for its further elucidation.
Collapse
Affiliation(s)
| | - Jonathan M Harnoss
- Department of General, Visceral, Thoracic and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Peter Walter
- Altos Labs, Inc., Bay Area Institute of Science, Redwood City, CA, USA.
| | - Avi Ashkenazi
- Research Oncology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
3
|
Yammine KM, Abularach SM, Xiong M, Kim SY, Bikovtseva AA, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, Shoulders MD. Human cartilage model of the precocious osteoarthritis-inducing COL2A1 p.Arg719Cys reveals pathology-driving matrix defects and a failure of the ER proteostasis network to recognize the defective procollagen-II. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622468. [PMID: 39574595 PMCID: PMC11580999 DOI: 10.1101/2024.11.07.622468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Objectives Mutations in the procollagen-II gene (COL2A1) often cause chondrodysplasias, including the precocious osteoarthritis-inducing p.Arg719Cys. Understanding the molecular basis of such diseases has long been challenging, owing to a lack of models accurately reflecting disease genotypes and phenotypes. To address this challenge, we develop and characterize in vitro human cartilage derived from wild-type and disease-causing Arg719Cys COL2A1 isogenic induced pluripotent stem cell (iPSC) lines. Methods Using directed differentiation of iPSCs to chondrocytes, we generated cartilage from wild-type and Arg719Cys COL2A1 lines. We compared the resulting protein, cell, and tissue properties using immunohistochemistry, electron microscopy, SDS-PAGE, RNA-sequencing, and quantitative interactomics. Results While both wild-type and disease lines deposited a cartilage matrix, the Arg719Cys matrix was deficient. Arg719Cys collagen-II was excessively post-translationally modified and modestly intracellularly retained, leading to endoplasmic reticulum (ER) distention suggestive of an ER storage defect. Interactomic studies indicated that Arg719Cys procollagen-II was not differentially engaged by the ER proteostasis network. RNA-sequencing showed that the ER storage defect engendered by Arg719Cys procollagen-II also did not activate cellular stress responses, including the unfolded protein response. These data suggest that cells fail to properly recognize Arg719Cys-associated procollagen-II defects. Conclusions A failure to identify and rectify defective procollagen-II folding in cells expressing Arg719Cys procollagen-II leads to the deposition of a sparse and defective collagen-II matrix, culminating in pathology. Combined with the highly expandable human cartilage disease model reported here, this work provides motivation and a platform to discover therapeutic strategies targeting procollagen folding, quality control, and secretion in this collagenopathy and others.
Collapse
Affiliation(s)
- Kathryn M Yammine
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sophia Mirda Abularach
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Michael Xiong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Seo-Yeon Kim
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Agata A Bikovtseva
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vincent L Butty
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Richard P Schiavoni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - John F Bateman
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Shireen R Lamandé
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
4
|
Zahoor M, Dong Y, Preussner M, Reiterer V, Shameen Alam S, Haun M, Horzum U, Frey Y, Hajdu R, Geley S, Cormier-Daire V, Heyd F, Jerome-Majewska LA, Farhan H. The unfolded protein response regulates ER exit sites via SNRPB-dependent RNA splicing and contributes to bone development. EMBO J 2024; 43:4228-4247. [PMID: 39160274 PMCID: PMC11445528 DOI: 10.1038/s44318-024-00208-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024] Open
Abstract
Splicing and endoplasmic reticulum (ER)-proteostasis are two key processes that ultimately regulate the functional proteins that are produced by a cell. However, the extent to which these processes interact remains poorly understood. Here, we identify SNRPB and other components of the Sm-ring, as targets of the unfolded protein response and novel regulators of export from the ER. Mechanistically, The Sm-ring regulates the splicing of components of the ER export machinery, including Sec16A, a component of ER exit sites. Loss of function of SNRPB is causally linked to cerebro-costo-mandibular syndrome (CCMS), a genetic disease characterized by bone defects. We show that heterozygous deletion of SNRPB in mice resulted in bone defects reminiscent of CCMS and that knockdown of SNRPB delays the trafficking of type-I collagen. Silencing SNRPB inhibited osteogenesis in vitro, which could be rescued by overexpression of Sec16A. This rescue indicates that the role of SNRPB in osteogenesis is linked to its effects on ER-export. Finally, we show that SNRPB is a target for the unfolded protein response, which supports a mechanistic link between the spliceosome and ER-proteostasis. Our work highlights components of the Sm-ring as a novel node in the proteostasis network, shedding light on CCMS pathophysiology.
Collapse
Affiliation(s)
- Muhammad Zahoor
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Yanchen Dong
- Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC, H4A 3J1, Canada
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Marco Preussner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Laboratory of RNA Biochemistry, Takustrasse 6, 14195, Berlin, Germany
| | - Veronika Reiterer
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Sabrina Shameen Alam
- Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC, H4A 3J1, Canada
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Margot Haun
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Utku Horzum
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Yannick Frey
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Renata Hajdu
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Stephan Geley
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Valerie Cormier-Daire
- Clinical Genetics Department, Université de Paris, INSERM UMR 1163, Imagine Institute, Necker Enfants Malades Hospital, Paris, France
| | - Florian Heyd
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Laboratory of RNA Biochemistry, Takustrasse 6, 14195, Berlin, Germany
| | - Loydie A Jerome-Majewska
- Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC, H4A 3J1, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 2B2, Canada.
- Department of Pediatrics, McGill University, Montreal, QC, H4A 3J1, Canada.
| | - Hesso Farhan
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
- Institute of Pathophysiology, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria.
| |
Collapse
|
5
|
Yammine KM, Mirda Abularach S, Kim SY, Bikovtseva AA, Lilianty J, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, Shoulders MD. ER procollagen storage defect without coupled unfolded protein response drives precocious arthritis. Life Sci Alliance 2024; 7:e202402842. [PMID: 38981683 PMCID: PMC11234256 DOI: 10.26508/lsa.202402842] [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: 05/28/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Likely owing to the unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific ER proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the easily expandable cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies.
Collapse
Affiliation(s)
- Kathryn M Yammine
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Seo-Yeon Kim
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Agata A Bikovtseva
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jinia Lilianty
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Vincent L Butty
- BioMicro Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Richard P Schiavoni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John F Bateman
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Shireen R Lamandé
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Australia
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
6
|
Yammine KM, Abularach SM, Kim SY, Bikovtseva AA, Lilianty J, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, Shoulders MD. ER procollagen storage defect without coupled unfolded protein response drives precocious arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.19.562780. [PMID: 37905055 PMCID: PMC10614947 DOI: 10.1101/2023.10.19.562780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Owing to unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies.
Collapse
|
7
|
Yu X, Ding H, Wang D, Ren Z, Chen B, Wu Q, Yuan T, Liu Y, Zhang L, Zhao J, Sun Z. Particle-induced osteolysis is mediated by endoplasmic reticulum stress-associated osteoblast apoptosis. Chem Biol Interact 2023; 383:110686. [PMID: 37659624 DOI: 10.1016/j.cbi.2023.110686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/29/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023]
Abstract
Osteoblast dysfunction plays a crucial role in periprosthetic osteolysis and aseptic loosening, and endoplasmic reticulum (ER) stress is recognized as an important causal factor of wear particle-induced osteolysis. However, the influence of ER stress on osteoblast activity during osteolysis and its underlying mechanisms remain elusive. This study aims to investigate whether ER stress is involved in the detrimental effects of wear particles on osteoblasts. Through our investigation, we observed elevated expression levels of ER stress and apoptosis markers in particle-stimulated bone specimens and osteoblasts. To probe further, we employed the ER stress inhibitor, 4-PBA, to treat particle-stimulated osteoblasts. The results revealed that 4-PBA effectively alleviated particle-induced osteoblast apoptosis and mitigated osteogenic reduction. Furthermore, our study revealed that wear particle-induced ER stress in osteoblasts coincided with mitochondrial damage, calcium overload, and oxidative stress, all of which were effectively alleviated by 4-PBA treatment. Encouragingly, 4-PBA administration also improved bone formation and attenuated osteolysis in a mouse calvarial model. In conclusion, our results demonstrate that ER stress plays a crucial role in mediating wear particle-induced osteoblast apoptosis and impaired osteogenic function. These findings underscore the critical involvement of ER stress in wear particle-induced osteolysis and highlight ER stress as a potential therapeutic target for ameliorating wear particle-induced osteogenic reduction and bone destruction.
Collapse
Affiliation(s)
- Xin Yu
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China
| | - Hao Ding
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China
| | - Dongsheng Wang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China
| | - Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210023, China
| | - Bin Chen
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China
| | - Qi Wu
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China
| | - Tao Yuan
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China
| | - Yang Liu
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710068, China.
| | - Lei Zhang
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China.
| | - Jianning Zhao
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China.
| | - Zhongyang Sun
- Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210093, China; Department of Orthopedics, Air Force Hospital of Eastern Theater, Anhui Medical University, Nanjing, 210002, China.
| |
Collapse
|
8
|
Wu T, Jiang Y, Shi W, Wang Y, Li T. Endoplasmic reticulum stress: a novel targeted approach to repair bone defects by regulating osteogenesis and angiogenesis. J Transl Med 2023; 21:480. [PMID: 37464413 PMCID: PMC10353205 DOI: 10.1186/s12967-023-04328-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Bone regeneration therapy is clinically important, and targeted regulation of endoplasmic reticulum (ER) stress is important in regenerative medicine. The processing of proteins in the ER controls cell fate. The accumulation of misfolded and unfolded proteins occurs in pathological states, triggering ER stress. ER stress restores homeostasis through three main mechanisms, including protein kinase-R-like ER kinase (PERK), inositol-requiring enzyme 1ɑ (IRE1ɑ) and activating transcription factor 6 (ATF6), collectively known as the unfolded protein response (UPR). However, the UPR has both adaptive and apoptotic effects. Modulation of ER stress has therapeutic potential for numerous diseases. Repair of bone defects involves both angiogenesis and bone regeneration. Here, we review the effects of ER stress on osteogenesis and angiogenesis, with emphasis on ER stress under high glucose (HG) and inflammatory conditions, and the use of ER stress inducers or inhibitors to regulate osteogenesis and angiogenesis. In addition, we highlight the ability for exosomes to regulate ER stress. Recent advances in the regulation of ER stress mediated osteogenesis and angiogenesis suggest novel therapeutic options for bone defects.
Collapse
Affiliation(s)
- Tingyu Wu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yaping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yingzhen Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China.
| |
Collapse
|
9
|
Iyer S, Adams DJ. Bone and the Unfolded Protein Response: In Sickness and in Health. Calcif Tissue Int 2023; 113:96-109. [PMID: 37243756 PMCID: PMC10326125 DOI: 10.1007/s00223-023-01096-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/08/2023] [Indexed: 05/29/2023]
Abstract
Differentiation and optimal function of osteoblasts and osteoclasts are contingent on synthesis and maintenance of a healthy proteome. Impaired and/or altered secretory capacity of these skeletal cells is a primary driver of most skeletal diseases. The endoplasmic reticulum (ER) orchestrates the folding and maturation of membrane as well as secreted proteins at high rates within a calcium rich and oxidative organellar niche. Three ER membrane proteins monitor fidelity of protein processing in the ER and initiate an intricate signaling cascade known as the Unfolded Protein Response (UPR) to remediate accumulation of misfolded proteins in its lumen, a condition referred to as ER stress. The UPR aids in fine-tuning, expanding and/or modifying the cellular proteome, especially in specialized secretory cells, to match everchanging physiologic cues and metabolic demands. Sustained activation of the UPR due to chronic ER stress, however, is known to hasten cell death and drive pathophysiology of several diseases. A growing body of evidence suggests that ER stress and an aberrant UPR may contribute to poor skeletal health and the development of osteoporosis. Small molecule therapeutics that target distinct components of the UPR may therefore have implications for developing novel treatment modalities relevant to the skeleton. This review summarizes the complexity of UPR actions in bone cells in the context of skeletal physiology and osteoporotic bone loss, and highlights the need for future mechanistic studies to develop novel UPR therapeutics that mitigate adverse skeletal outcomes.
Collapse
Affiliation(s)
- Srividhya Iyer
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Mailstop:8343, Aurora, CO, 80045, USA.
| | - Douglas J Adams
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Mailstop:8343, Aurora, CO, 80045, USA
| |
Collapse
|
10
|
Huang W, Gong Y, Yan L. ER Stress, the Unfolded Protein Response and Osteoclastogenesis: A Review. Biomolecules 2023; 13:1050. [PMID: 37509086 PMCID: PMC10377020 DOI: 10.3390/biom13071050] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Endoplasmic reticulum (ER) stress and its adaptive mechanism, the unfolded protein response (UPR), are triggered by the accumulation of unfolded and misfolded proteins. During osteoclastogenesis, a large number of active proteins are synthesized. When an imbalance in the protein folding process occurs, it causes osteoclasts to trigger the UPR. This close association has led to the role of the UPR in osteoclastogenesis being increasingly explored. In recent years, several studies have reported the role of ER stress and UPR in osteoclastogenesis and bone resorption. Here, we reviewed the relevant literature and discussed the UPR signaling cascade response, osteoclastogenesis-related signaling pathways, and the role of UPR in osteoclastogenesis and bone resorption in detail. It was found that the UPR signal (PERK, CHOP, and IRE1-XBP1) promoted the expression of the receptor activator of the nuclear factor-kappa B ligand (RANKL) in osteoblasts and indirectly enhanced osteoclastogenesis. IRE1 promoted osteoclastogenesis via promoting NF-κB, MAPK signaling, or the release of pro-inflammatory factors (IL-6, IL-1β, and TNFα). CREBH promoted osteoclast differentiation by promoting NFATc1 expression. The PERK signaling pathway also promoted osteoclastogenesis through NF-κB and MAPK signaling pathways, autophagy, and RANKL secretion from osteoblasts. However, salubrinal (an inhibitor of eIF2α dephosphorylation that upregulated p-eIF2α expression) directly inhibited osteoclastogenesis by suppressing NFATc1 expression and indirectly promoted osteoclastogenesis by promoting RANKL secretion from osteoblasts. Therefore, the specific effects and mechanisms of p-PERK and its downstream signaling on osteoclastogenesis still need further experiments to confirm. In addition, the exact role of ATF6 and BiP in osteoclastogenesis also required further exploration. In conclusion, our detailed and systematic review provides some references for the next step to fully elucidate the relationship between UPR and osteoclastogenesis, intending to provide new insights for the treatment of diseases caused by osteoclast over-differentiation, such as osteoporosis.
Collapse
Affiliation(s)
- Wangli Huang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yining Gong
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Liang Yan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| |
Collapse
|
11
|
Fu W, Im YG, Kim B, Kim OS, Yang Y, Song J, Liu D, Zhu S, Kang JS, Kim O. 625 nm Light Irradiation Prevented MC3T3-E1 Cells from Accumulation of Misfolded Proteins via ROS and ATP Production. Int J Mol Sci 2023; 24:ijms24119257. [PMID: 37298212 DOI: 10.3390/ijms24119257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Osteoblasts must acquire a considerable capacity for folding unfolded and misfolded proteins (MPs) to produce large amounts of extracellular matrix proteins and maintain bone homeostasis. MP accumulation contributes to cellular apoptosis and bone disorders. Photobiomodulation therapy has been used to treat bone diseases, but the effects of decreasing MPs with photobiomodulation remain unclear. In this study, we explored the efficacy of 625 nm light-emitting diode irradiation (LEDI) to reduce MPs in tunicamycin (TM) induced-MC3T3-E1 cells. Binding immunoglobulin protein (BiP), an adenosine triphosphate (ATP)-dependent chaperone, is used to evaluate the capacity of folding MPs. The results revealed that pretreatment with 625 nm LEDI (Pre-IR) induced reactive oxygen species (ROS) production, leading to the increased chaperone BiP through the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1s (XBP-1s) pathway, and then restoration of collagen type I (COL-I) and osteopontin (OPN) expression relieving cell apoptosis. Furthermore, the translocation of BiP into the endoplasmic reticulum (ER) lumen might be followed by a high level of ATP production. Taken together, these results suggest that Pre-IR could be beneficial to prevent MP accumulation through ROS and ATP in TM-induced MC3T3-E1cells.
Collapse
Affiliation(s)
- Wenqi Fu
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yeong-Gwan Im
- Department of Oral Medicine, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Byunggook Kim
- Department of Oral Medicine, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ok-Su Kim
- Department of Periodontology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ying Yang
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jianan Song
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Danyang Liu
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Siyu Zhu
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jae-Seok Kang
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Okjoon Kim
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
12
|
Salazar-Noratto GE, Nations CC, Stevens HY, Xu M, Gaynard S, Dooley C, de Nijs N, McDonagh K, Shen S, Willimon SC, Barry F, Guldberg RE. Patient-Specific iPSC-Derived Models Link Aberrant Endoplasmic Reticulum Stress Sensing and Response to Juvenile Osteochondritis Dissecans Etiology. Stem Cells Transl Med 2023; 12:293-306. [PMID: 37184892 PMCID: PMC10184700 DOI: 10.1093/stcltm/szad018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/19/2023] [Indexed: 05/16/2023] Open
Abstract
Juvenile osteochondritis dissecans (JOCD) is a pediatric disease, which begins with an osteonecrotic lesion in the secondary ossification center which, over time, results in the separation of the necrotic fragment from the parent bone. JOCD predisposes to early-onset osteoarthritis. However, the knowledge gap in JOCD pathomechanisms severely limits current therapeutic strategies. To elucidate its etiology, we conducted a study with induced pluripotent stem cells (iPSCs) from JOCD and control patients. iPSCs from skin biopsies were differentiated to iMSCs (iPSC-derived mesenchymal stromal cells) and subjected to chondrogenic and endochondral ossification, and endoplasmic reticulum (ER)-stress induction assays. Our study, using 3 JOCD donors, showed that JOCD cells have lower chondrogenic capability and their endochondral ossification process differs from control cells; yet, JOCD- and control-cells accomplish osteogenesis of similar quality. Our findings show that endoplasmic reticulum stress sensing and response mechanisms in JOCD cells, which partially regulate chondrocyte and osteoblast differentiation, are related to these differences. We suggest that JOCD cells are more sensitive to ER stress than control cells, and in pathological microenvironments, such as microtrauma and micro-ischemia, JOCD pathogenesis pathways may be initiated. This study is the first, to the best of our knowledge, to realize the important role that resident cells and their differentiating counterparts play in JOCD and to put forth a novel etiological hypothesis that seeks to consolidate and explain previously postulated hypotheses. Furthermore, our results establish well-characterized JOCD-specific iPSC-derived in vitro models and identified potential targets which could be used to improve diagnostic tools and therapeutic strategies in JOCD.
Collapse
Affiliation(s)
- Giuliana E Salazar-Noratto
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Catriana C Nations
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hazel Y Stevens
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Maojia Xu
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Sean Gaynard
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Claire Dooley
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Nica de Nijs
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Katya McDonagh
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - S Clifton Willimon
- Children's Orthopaedics of Atlanta, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Frank Barry
- Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Robert E Guldberg
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
13
|
Sel1l May Contributes to the Determinants of Neuronal Lineage and Neuronal Maturation Regardless of Hrd1 via Atf6-Sel1l Signaling. Neurochem Res 2023; 48:263-272. [PMID: 36074198 DOI: 10.1007/s11064-022-03750-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/08/2022] [Accepted: 08/30/2022] [Indexed: 01/11/2023]
Abstract
The endoplasmic reticulum (ER) is the primary site of intracellular quality control involved in the recognition and degradation of unfolded proteins. A variety of stresses, including hypoxia and glucose starvation, can lead to accumulation of unfolded proteins triggering the ER-associated degradation (ERAD) pathway. Suppressor Enhancer Lin12/Notch1 Like (Sel1l) acts as a "gate keeper" in the quality control of de novo synthesized proteins and complexes with the ubiquitin ligase Hrd1 in the ER membrane. We previously demonstrated that ER stress-induced aberrant neural stem cell (NSC) differentiation and inhibited neurite outgrowth. Inhibition of neurite outgrowth was associated with increased Hrd1 expression; however, the contribution of Sel1l remained unclear. To investigate whether ER stress is induced during normal neuronal differentiation, we semi-quantitatively evaluated mRNA expression levels of unfolded protein response (UPR)-related genes in P19 embryonic carcinoma cells undergoing neuronal differentiation in vitro. Stimulation with all-trans retinoic acid (ATRA) for 4 days induced the upregulation of Nestin and several UPR-related genes (Atf6, Xbp1, Chop, Hrd1, and Sel1l), whereas Atf4 and Grp78/Bip were unchanged. Small-interfering RNA (siRNA)-mediated knockdown of Sel1l uncovered that mRNA levels of the neural progenitor marker Math1 (also known as Atoh1) and the neuronal marker Math3 (also known as Atoh3 and NeuroD4) were significantly suppressed at 4 days after ATRA stimulation. Consistent with this result, Sel1l silencing significantly reduced protein levels of immature neuronal marker βIII-tubulin (also known as Tuj-1) at 8 days after induction of neuronal differentiation, whereas synaptogenic factors, such as cell adhesion molecule 1 (CADM1) and SH3 and multiple ankyrin repeat domain protein 3 (Shank3) were accumulated in Sel1l silenced cells. These results indicate that neuronal differentiation triggers ER stress and suggest that Sel1l may facilitate neuronal lineage through the regulation of Math1 and Math3 expression.
Collapse
|
14
|
Yu X, Xu X, Dong W, Yang C, Luo Y, He Y, Jiang C, Wu Y, Wang J. DDIT3/CHOP mediates the inhibitory effect of ER stress on chondrocyte differentiation by AMPKα-SIRT1 pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119265. [PMID: 35381294 DOI: 10.1016/j.bbamcr.2022.119265] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Endoplasmic reticulum (ER) stress is an evolutionarily conserved cellular stress response related to multiple diseases, including temporomandibular joint (TMJ) cartilage-related diseases. Recent studies have indicated that DDIT3/CHOP (a downstream transcription factor of ER stress) is an important effector in mediating ER stress to inhibit chondrogenesis. However, the underlying mechanism by which DDIT3 regulates chondrogenesis remains unclear. In this study, tunicamycin (an ER stress agonist)-induced ER stress inhibited chondrocyte differentiation and matrix synthesis in vitro and led to an osteoarthritis-like phenotype in mouse TMJ cartilage. Meanwhile, DDIT3 expression in chondrocytes was robustly upregulated. Loss-of-function experiments validated the inhibiting effect of DDIT3 on chondrocyte differentiation and matrix synthesis. Mechanistically, the inhibiting effect was attributed to the direct and indirect regulatory effect of DDIT3 on SIRT1 (sirtuin1, silent mating type information regulation protein type 1, a member of NAD+ dependent class III histone deacetylases). On one hand, DDIT3 directly promoted the transcription of SIRT1. On the other hand, DDIT3 indirectly increased the expression of SIRT1 by promoting AMPKα phosphorylation and activation. Furthermore, activation of AMPKα or SIRT1 with the corresponding agonist AICAR or resveratrol in the DDIT3-knockdown cells partially restored the inhibiting effect of DDIT3 on chondrocyte differentiation and matrix synthesis. Collectively, these novel findings indicate that DDIT3 regulates the inhibitory effect of ER stress on chondrocyte differentiation and matrix synthesis partially via the AMPKα-SIRT1 pathway. A thorough understanding of ER stress in regulating chondrocyte homeostasis and its role in the onset of osteoarthritis may be promising to develop therapeutic targets and prevent condyle cartilage destruction.
Collapse
Affiliation(s)
- Xijie Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xiaoxiao Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Dong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Yao Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Ying He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chenxi Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yanru Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jiawei Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
15
|
METTL3 mediates osteoblast apoptosis by regulating endoplasmic reticulum stress during LPS-induced inflammation. Cell Signal 2022; 95:110335. [DOI: 10.1016/j.cellsig.2022.110335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022]
|
16
|
Wood RK, Flory AR, Mann MJ, Talbot LJ, Hendershot LM. Secretory defects in pediatric osteosarcoma result from downregulation of selective COPII coatomer proteins. iScience 2022; 25:104100. [PMID: 35402877 PMCID: PMC8983387 DOI: 10.1016/j.isci.2022.104100] [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: 10/05/2021] [Revised: 01/11/2022] [Accepted: 03/15/2022] [Indexed: 12/05/2022] Open
Abstract
Pediatric osteosarcomas (OS) exhibit extensive genomic instability that has complicated the identification of new targeted therapies. We found the vast majority of 108 patient tumor samples and patient-derived xenografts (PDXs), which display an unusually dilated endoplasmic reticulum (ER), have reduced expression of four COPII vesicle components that trigger aberrant accumulation of procollagen-I protein within the ER. CRISPR activation technology was used to increase the expression of two of these, SAR1A and SEC24D, to physiological levels. This was sufficient to resolve the dilated ER morphology, restore collagen-I secretion, and enhance secretion of some extracellular matrix (ECM) proteins. However, orthotopic xenograft growth was not adversely affected by restoration of only SAR1A and SEC24D. Our studies reveal the mechanism responsible for the dilated ER that is a hallmark characteristic of OS and identify a highly conserved molecular signature for this genetically unstable tumor. Possible relationships of this phenotype to tumorigenesis are discussed.
Collapse
Affiliation(s)
- Rachael K. Wood
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ashley R. Flory
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Melissa J. Mann
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lindsay J. Talbot
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
17
|
Osteogenic Commitment of Human Periodontal Ligament Cells Is Predetermined by Methylation, Chromatin Accessibility and Expression of Key Transcription Factors. Cells 2022; 11:cells11071126. [PMID: 35406691 PMCID: PMC8997528 DOI: 10.3390/cells11071126] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Periodontal ligament stem cells (PDLCs) can be used as a valuable source in cell therapies to regenerate bone tissue. However, the potential therapeutic outcomes are unpredictable due to PDLCs’ heterogeneity regarding the capacity for osteoblast differentiation and mineral nodules production. Here, we identify epigenetic (DNA (hydroxy)methylation), chromatin (ATAC-seq) and transcriptional (RNA-seq) differences between PDLCs presenting with low (l) and high (h) osteogenic potential. The primary cell populations were investigated at basal state (cultured in DMEM) and after 10 days of osteogenic stimulation (OM). At a basal state, the expression of transcription factors (TFs) and the presence of gene regulatory regions related to osteogenesis were detected in h-PDLCs in contrast to neuronal differentiation prevalent in l-PDLCs. These differences were also observed under stimulated conditions, with genes and biological processes associated with osteoblast phenotype activated more in h-PDLCs. Importantly, even after the induction, l-PDLCs showed hypermethylation and low expression of genes related to bone development. Furthermore, the analysis of TFs motifs combined with TFs expression suggested the relevance of SP1, SP7 and DLX4 regulation in h-PDLCs, while motifs for SIX and OLIG2 TFs were uniquely enriched in l-PDLCs. Additional analysis including a second l-PDLC population indicated that the high expression of OCT4, SIX3 and PPARG TFs could be predictive of low osteogenic commitment. In summary, several biological processes related to osteoblast commitment were activated in h-PDLCs from the onset, while l-PDLCs showed delay in the activation of the osteoblastic program, restricted by the persistent methylation of gene related to bone development. These processes are pre-determined by distinguishable epigenetic and transcriptional patterns, the recognition of which could help in selection of PDLCs with pre-osteoblastic phenotype.
Collapse
|
18
|
Yang L, Li Z, Ouyang Y. Taurine attenuates ER stress‑associated apoptosis and catabolism in nucleus pulposus cells. Mol Med Rep 2022; 25:172. [PMID: 35315493 PMCID: PMC8971911 DOI: 10.3892/mmr.2022.12688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 11/20/2022] Open
Abstract
Nucleus pulposus (NP) apoptosis and subsequent excessive degradation of the extracellular matrix (ECM) are key pathological characteristics of intervertebral disc degeneration (IDD). The present study aims to examine the signaling processes underlying the effects of taurine on IDD, with specific focus on endoplasmic reticulum (ER) stress-mediated apoptosis and ECM degradation, in NP cells. To clarify the role of taurine in IDD, NP cells were treated with various concentrations of taurine and IL-1β or thapsigargin (TG). Cell Counting Kit-8, western blotting, TUNEL, immunofluorescence assays and reverse transcription-quantitative PCR were applied to measure cell viability, the expression of ER stress-associated proteins (GRP78, CHOP and caspase-12), apoptosis and the levels of metabolic factors associated with ECM (MMP-1, 3, 9, ADAMTS-4, 5 and collagen II), respectively. Taurine was found to attenuate ER stress and prevent apoptosis in NP cells induced by IL-1β treatment. Additionally, taurine significantly decreased the expression of ER stress-activated glucose regulatory protein 78, C/EBP homologous protein and caspase-12. TUNEL results revealed that taurine decreased the number of apoptotic TG-treated NP cells. TG-treated NP cells also exhibited characteristics of increased ECM degradation, supported by observations of increased MMP-1, MMP-3, MMP-9 and A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 expression in addition to decreased collagen-II expression. However, taurine treatment significantly reversed all indicators of excessive ECM catabolism aforementioned. These data suggest that taurine can mediate protection against apoptosis and ECM degradation in NP cells by inhibiting ER stress, implicating therapeutic potential for the treatment of IDD.
Collapse
Affiliation(s)
- Liuxie Yang
- Department of Orthopedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200040, P.R. China
| | - Zhenhuan Li
- Department of Orthopedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200040, P.R. China
| | - Yueping Ouyang
- Department of Orthopedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
19
|
Echinacoside Upregulates Sirt1 to Suppress Endoplasmic Reticulum Stress and Inhibit Extracellular Matrix Degradation In Vitro and Ameliorates Osteoarthritis In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3137066. [PMID: 34777682 PMCID: PMC8580641 DOI: 10.1155/2021/3137066] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/08/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022]
Abstract
Background Osteoarthritis (OA) is a progressive illness that destroys cartilage. Oxidative stress is a major contributor of OA, while endoplasmic reticulum (ER) stress is the key cellular damage under oxidative stress in chondrocytes. Echinacoside (ECH) is the main extract and active substance of Cistanche, with potent antioxidative stress (OS) properties, and currently under clinical trials in China. However, its function in OA is yet to be determined. Purpose We aimed to explore the specific role of ECH in the occurrence and development of OA and its underlying mechanism in vivo and in vitro. Methods After the mice were anesthetized, the bilateral medial knee joint meniscus resection was performed to establish the DMM model. TBHP was used to induce oxidative stress to establish the OA model in chondrocytes in vitro. Western blot and RT-PCR were used to evaluate the level of ER stress-related biomarkers such as p-PERK/PERK, GRP78, ATF4, p-eIF2α/eIF2α, and CHOP and apoptosis-related proteins such as BAX, Bcl-2, and cleaved caspase-3. Meanwhile, we used SO staining, immunofluorescence, and immunohistochemical staining to evaluate the pharmacological effects of ECH in mice in vivo. Results We demonstrated the effectiveness of ECH in suppressing ER stress and restoring ECM metabolism in vitro. In particular, ECH was shown to suppress tert-Butyl hydroperoxide- (TBHP-) induced OS and subsequently lower the levels of p-PERK/PERK, GRP78, ATF4, p-eIF2α/eIF2α, and CHOP in vitro. Simultaneously, ECH reduced MMP13 and ADAMTS5 levels and promoted Aggrecan and Collagen II levels, suggesting ECM degradation suppression. Moreover, we showed that ECH mediates its cellular effects via upregulation of Sirt1. Lastly, we confirmed that ECH can protect against OA in mouse OA models. Conclusion In summary, our findings indicate that ECH can inhibit ER stress and ECM degradation by upregulating Sirt1 in mouse chondrocytes treated with TBHP. It can also prevent OA development in vivo.
Collapse
|
20
|
Wen C, Zhou Y, Xu Y, Tan H, Pang C, Liu H, Liu K, Wei L, Luo H, Qin T, He C, Liu C, Zhou C. The Regulatory Role of GBF1 on Osteoclast Activation Through EIF2a Mediated ER Stress and Novel Marker FAM129A Induction. Front Cell Dev Biol 2021; 9:706768. [PMID: 34513838 PMCID: PMC8424197 DOI: 10.3389/fcell.2021.706768] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022] Open
Abstract
Bone-resorbing activities of osteoclasts (OCs) are highly dependent on actin cytoskeleton remodeling, plasma membrane reorganization, and vesicle trafficking pathways, which are partially regulated by ARF-GTPases. In the present study, the functional roles of Golgi brefeldin A resistance factor 1 (GBF1) are proposed. GBF1 is responsible for the activation of the ARFs family and vesicular transport at the endoplasmic reticulum–Golgi interface in different stages of OCs differentiation. In the early stage, GBF1 deficiency impaired OCs differentiation and was accompanied with OCs swelling and reduced formation of mature OCs, indicating that GBF1 participates in osteoclastogenesis. Using siRNA and the specific inhibitor GCA for GBF1 knockdown upregulated endoplasmic reticulum stress-associated signaling molecules, including BiP, p-PERK, p-EIF2α, and FAM129A, and promoted autophagic Beclin1, Atg7, p62, and LC3 axis, leading to apoptosis of OCs. The present data suggest that, by blocking COPI-mediated vesicular trafficking, GBF1 inhibition caused intense stress to the endoplasmic reticulum and excessive autophagy, eventually resulting in the apoptosis of mature OCs and impaired bone resorption function.
Collapse
Affiliation(s)
- Cailing Wen
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuheng Zhou
- School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Yanting Xu
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Huijing Tan
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Caixia Pang
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Haiqian Liu
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Kaifei Liu
- Department of Pharmacy, Jingzhou Central Hospital, Jingzhou, China
| | - Linlin Wei
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Luo
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Tian Qin
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chonghua He
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Cuiling Liu
- Shenzhen Bao'an District Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Chun Zhou
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Liang D, Khoonkari M, Avril T, Chevet E, Kruyt FAE. The unfolded protein response as regulator of cancer stemness and differentiation: Mechanisms and implications for cancer therapy. Biochem Pharmacol 2021; 192:114737. [PMID: 34411568 DOI: 10.1016/j.bcp.2021.114737] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022]
Abstract
The unfolded protein response (UPR) is an adaptive mechanism that regulates protein and cellular homeostasis. Three endoplasmic reticulum (ER) membrane localized stress sensors, IRE1, PERK and ATF6, coordinate the UPR in order to maintain ER proteostasis and cell survival, or induce cell death when homeostasis cannot be restored. However, recent studies have identified alternative functions for the UPR in developmental biology processes and cell fate decisions under both normal and cancerous conditions. In cancer, increasing evidence points towards the involvement of the three UPR sensors in oncogenic reprogramming and the regulation of tumor cells endowed with stem cell properties, named cancer stem cells (CSCs), that are considered to be the most malignant cells in tumors. Here we review the reported roles and underlying molecular mechanisms of the three UPR sensors in regulating stemness and differentiation, particularly in solid tumor cells, processes that have a major impact on tumor aggressiveness. Mainly PERK and IRE1 branches of the UPR were found to regulate CSCs and tumor development and examples are provided for breast cancer, colon cancer and aggressive brain tumors, glioblastoma. Although the underlying mechanisms and interactions between the different UPR branches in regulating stemness in cancer need to be further elucidated, we propose that PERK and IRE1 targeted therapy could inhibit self-renewal of CSCs or induce differentiation that is predicted to have therapeutic benefit. For this, more specific UPR modulators need to be developed with favorable pharmacological properties that together with patient stratification will allow optimal evaluation in clinical studies.
Collapse
Affiliation(s)
- Dong Liang
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Mohammad Khoonkari
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Tony Avril
- INSERM U1242, Université de Rennes, Rennes, France; Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Eric Chevet
- INSERM U1242, Université de Rennes, Rennes, France; Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
22
|
Collagen's enigmatic, highly conserved N-glycan has an essential proteostatic function. Proc Natl Acad Sci U S A 2021; 118:2026608118. [PMID: 33674390 DOI: 10.1073/pnas.2026608118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Intracellular procollagen folding begins at the protein's C-terminal propeptide (C-Pro) domain, which initiates triple-helix assembly and defines the composition and chain register of fibrillar collagen trimers. The C-Pro domain is later proteolytically cleaved and excreted from the body, while the mature triple helix is incorporated into the extracellular matrix. The procollagen C-Pro domain possesses a single N-glycosylation site that is widely conserved in all the fibrillar procollagens across humans and diverse other species. Given that the C-Pro domain is removed once procollagen folding is complete, the N-glycan might be presumed to be important for folding. Surprisingly, however, there is no difference in the folding and secretion of N-glycosylated versus non-N-glycosylated collagen type-I, leaving the function of the N-glycan unclear. We hypothesized that the collagen N-glycan might have a context-dependent function, specifically, that it could be required to promote procollagen folding only when proteostasis is challenged. We show that removal of the N-glycan from misfolding-prone C-Pro domain variants does indeed cause serious procollagen and ER proteostasis defects. The N-glycan promotes folding and secretion of destabilized C-Pro variants by providing access to the ER's lectin-based chaperone machinery. Finally, we show that the C-Pro N-glycan is actually critical for the folding and secretion of even wild-type procollagen under ER stress conditions. Such stress is commonly incurred during development, wound healing, and other processes in which collagen production plays a key role. Collectively, these results establish an essential, context-dependent function for procollagen's previously enigmatic N-glycan, wherein the carbohydrate moiety buffers procollagen folding against proteostatic challenge.
Collapse
|
23
|
Garibaldi N, Contento BM, Babini G, Morini J, Siciliani S, Biggiogera M, Raspanti M, Marini JC, Rossi A, Forlino A, Besio R. Targeting cellular stress in vitro improves osteoblast homeostasis, matrix collagen content and mineralization in two murine models of osteogenesis imperfecta. Matrix Biol 2021; 98:1-20. [PMID: 33798677 PMCID: PMC11162743 DOI: 10.1016/j.matbio.2021.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022]
Abstract
Most cases of dominantly inherited osteogenesis imperfecta (OI) are caused by glycine substitutions in the triple helical domain of type I collagen α chains, which delay collagen folding, and cause the synthesis of collagen triple helical molecules with abnormal structure and post-translational modification. A variable extent of mutant collagen ER retention and other secondary mutation effects perturb osteoblast homeostasis and impair bone matrix quality. Amelioration of OI osteoblast homeostasis could be beneficial both to osteoblast anabolic activity and to the content of the extracellular matrix they deposit. Therefore, the effect of the chemical chaperone 4-phenylbutyrate (4-PBA) on cell homeostasis, collagen trafficking, matrix production and mineralization was investigated in primary osteoblasts from two murine models of moderate OI, Col1a1+/G349C and Col1a2+/G610C. At the cellular level, 4-PBA prevented intracellular accumulation of collagen and increased protein secretion, reducing aggregates within the mutant cells and normalizing ER morphology. At the extracellular level, increased collagen incorporation into matrix, associated with more mature collagen fibrils, was observed in osteoblasts from both models. 4-PBA also promoted OI osteoblast mineral deposition by increasing alkaline phosphatase expression and activity. Targeting osteoblast stress with 4-PBA improved both cellular and matrix abnormalities in culture, supporting further in vivo studies of its effect on bone tissue composition, strength and mineralization as a potential treatment for classical OI.
Collapse
Affiliation(s)
- Nadia Garibaldi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy; Istituto Universitario di Studi Superiori - IUSS, Pavia, Italy.
| | - Barbara M Contento
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | | | - Jacopo Morini
- Department of Physics, University of Pavia, Pavia, Italy.
| | - Stella Siciliani
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.
| | - Marco Biggiogera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.
| | - Mario Raspanti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Joan C Marini
- Bone and Extracellular Matrix Branch, NICHD, National Institute of Health, Bethesda, MD 20892, USA.
| | - Antonio Rossi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| |
Collapse
|
24
|
Guo J, Ren R, Sun K, He J, Shao J. PERK signaling pathway in bone metabolism: Friend or foe? Cell Prolif 2021; 54:e13011. [PMID: 33615575 PMCID: PMC8016635 DOI: 10.1111/cpr.13011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/18/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoblasts and osteoclasts participate in the process of bone remodelling to meet the needs of normal growth and development or repair pathological damage. Endoplasmic reticulum stress (ER stress) can break the intracellular homeostasis of osteoclasts and osteoblasts, which is closely related to abnormal bone remodelling. The double‐stranded RNA‐dependent protein kinase (PKR)‐like ER kinase (PERK) is a key transmembrane protein that regulates ER stress, and growing evidence suggests that the PERK pathway plays a crucial role in regulating bone metabolism under both physiological and pathological conditions. Based on the current findings, we summarized the main mechanisms involved in bone metabolism downstream of the PERK pathway, among which elF2α, FOXO1, CaN, Nrf2 and DAG play a role in regulating the differentiation of osteoblasts and osteoclasts. Importantly, strategies by the regulation of PERK pathway exert beneficial effects in preclinical trials of several bone‐related diseases. Given the importance and novelty of PERK pathway, we provide an overview and discuss the roles of PERK pathway in regulating bone metabolism and its impact on bone‐related diseases. We hope that the development of research in this field will bring a bright future for the treatment of bone‐related diseases.
Collapse
Affiliation(s)
- Jiachao Guo
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ranyue Ren
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinpeng He
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingfan Shao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Dennis EP, Greenhalgh-Maychell PL, Briggs MD. Multiple epiphyseal dysplasia and related disorders: Molecular genetics, disease mechanisms, and therapeutic avenues. Dev Dyn 2020; 250:345-359. [PMID: 32633442 DOI: 10.1002/dvdy.221] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
For the vast majority of the 6000 known rare disease the pathogenic mechanisms are poorly defined and there is little treatment, leading to poor quality of life and high healthcare costs. Genetic skeletal diseases (skeletal dysplasias) are archetypal examples of rare diseases that are chronically debilitating, often life-threatening and for which no treatments are currently available. There are more than 450 unique phenotypes that, although individually rare, have an overall prevalence of at least 1 per 4000 children. Multiple epiphyseal dysplasia (MED) is a clinically and genetically heterogeneous disorder characterized by disproportionate short stature, joint pain, and early-onset osteoarthritis. MED is caused by mutations in the genes encoding important cartilage extracellular matrix proteins, enzymes, and transporter proteins. Recently, through the use of various cell and mouse models, disease mechanisms underlying this diverse phenotypic spectrum are starting to be elucidated. For example, ER stress induced as a consequence of retained misfolded mutant proteins has emerged as a unifying disease mechanisms for several forms of MED in particular and skeletal dysplasia in general. Moreover, targeting ER stress through drug repurposing has become an attractive therapeutic avenue.
Collapse
Affiliation(s)
- Ella P Dennis
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| | | | - Michael D Briggs
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| |
Collapse
|
26
|
Abstract
Cartilage comprises a single cell type, the chondrocyte, embedded in a highly complex extracellular matrix. Disruption to the cartilage growth plate leads to reduced bone growth and results in a clinically diverse group of conditions known as genetic skeletal diseases (GSDs). Similarly, long-term degradation of articular cartilage can lead to osteoarthritis (OA), a disease characterised by joint pain and stiffness. As professionally secreting cells, chondrocytes are particularly susceptible to endoplasmic reticulum (ER) stress and this has been identified as a core disease mechanism in a group of clinically and pathologically related GSDs. If unresolved, ER stress can lead to chondrocyte cell death. Recent interest has focused on ER stress as a druggable target for GSDs and this has led to the first clinical trial for a GSD by repurposing an antiepileptic drug. Interestingly, ER stress markers have also been associated with OA in multiple cell and animal models and there is increasing interest in it as a possible therapeutic target for treatment. In summary, chondrocyte ER stress has been identified as a core disease mechanism in GSDs and as a contributory factor in OA. Thus, chondrocyte ER stress is a unifying factor for both common and rare cartilage-related diseases and holds promise as a novel therapeutic target.
Collapse
Affiliation(s)
- Michael D Briggs
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Ella P Dennis
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Helen F Dietmar
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Katarzyna A Pirog
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
27
|
Sledgehammer to Scalpel: Broad Challenges to the Heart and Other Tissues Yield Specific Cellular Responses via Transcriptional Regulation of the ER-Stress Master Regulator ATF6α. Int J Mol Sci 2020; 21:ijms21031134. [PMID: 32046286 PMCID: PMC7037772 DOI: 10.3390/ijms21031134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/28/2022] Open
Abstract
There are more than 2000 transcription factors in eukaryotes, many of which are subject to complex mechanisms fine-tuning their activity and their transcriptional programs to meet the vast array of conditions under which cells must adapt to thrive and survive. For example, conditions that impair protein folding in the endoplasmic reticulum (ER), sometimes called ER stress, elicit the relocation of the ER-transmembrane protein, activating transcription factor 6α (ATF6α), to the Golgi, where it is proteolytically cleaved. This generates a fragment of ATF6α that translocates to the nucleus, where it regulates numerous genes that restore ER protein-folding capacity but is degraded soon after. Thus, upon ER stress, ATF6α is converted from a stable, transmembrane protein, to a rapidly degraded, nuclear protein that is a potent transcription factor. This review focuses on the molecular mechanisms governing ATF6α location, activity, and stability, as well as the transcriptional programs ATF6α regulates, whether canonical genes that restore ER protein-folding or unexpected, non-canonical genes affecting cellular functions beyond the ER. Moreover, we will review fascinating roles for an ATF6α isoform, ATF6β, which has a similar mode of activation but, unlike ATF6α, is a long-lived, weak transcription factor that may moderate the genetic effects of ATF6α.
Collapse
|
28
|
Martelli AM, Paganelli F, Chiarini F, Evangelisti C, McCubrey JA. The Unfolded Protein Response: A Novel Therapeutic Target in Acute Leukemias. Cancers (Basel) 2020; 12:cancers12020333. [PMID: 32024211 PMCID: PMC7072709 DOI: 10.3390/cancers12020333] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive response triggered by the stress of the endoplasmic reticulum (ER) due, among other causes, to altered cell protein homeostasis (proteostasis). UPR is mediated by three main sensors, protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 6α (ATF6α), and inositol-requiring enzyme-1α (IRE1α). Given that proteostasis is frequently disregulated in cancer, UPR is emerging as a critical signaling network in controlling the survival, selection, and adaptation of a variety of neoplasias, including breast cancer, prostate cancer, colorectal cancer, and glioblastoma. Indeed, cancer cells can escape from the apoptotic pathways elicited by ER stress by switching UPR into a prosurvival mechanism instead of cell death. Although most of the studies on UPR focused on solid tumors, this intricate network plays a critical role in hematological malignancies, and especially in multiple myeloma (MM), where treatment with proteasome inhibitors induce the accumulation of unfolded proteins that severely perturb proteostasis, thereby leading to ER stress, and, eventually, to apoptosis. However, UPR is emerging as a key player also in acute leukemias, where recent evidence points to the likelihood that targeting UPR-driven prosurvival pathways could represent a novel therapeutic strategy. In this review, we focus on the oncogene-specific regulation of individual UPR signaling arms, and we provide an updated outline of the genetic, biochemical, and preclinical therapeutic findings that support UPR as a relevant, novel target in acute leukemias.
Collapse
Affiliation(s)
- Alberto M. Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy;
- Correspondence: ; Tel.: +39-051-209-1580
| | - Francesca Paganelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Francesca Chiarini
- CNR Institute of Molecular Genetics, 40136 Bologna, Italy; (F.C.); (C.E.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Camilla Evangelisti
- CNR Institute of Molecular Genetics, 40136 Bologna, Italy; (F.C.); (C.E.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - James A. McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| |
Collapse
|
29
|
Li J, Li X, Liu D, Hamamura K, Wan Q, Na S, Yokota H, Zhang P. eIF2α signaling regulates autophagy of osteoblasts and the development of osteoclasts in OVX mice. Cell Death Dis 2019; 10:921. [PMID: 31801950 PMCID: PMC6892793 DOI: 10.1038/s41419-019-2159-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022]
Abstract
Bone loss in postmenopausal osteoporosis is induced chiefly by an imbalance of bone-forming osteoblasts and bone-resorbing osteoclasts. Salubrinal is a synthetic compound that inhibits de-phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α). Phosphorylation of eIF2α alleviates endoplasmic reticulum (ER) stress, which may activate autophagy. We hypothesized that eIF2α signaling regulates bone homeostasis by promoting autophagy in osteoblasts and inhibiting osteoclast development. To test the hypothesis, we employed salubrinal to elevate the phosphorylation of eIF2α in an ovariectomized (OVX) mouse model and cell cultures. In the OVX model, salubrinal prevented abnormal expansion of rough ER and decreased the number of acidic vesiculars. It regulated ER stress-associated signaling molecules such as Bip, p-eIF2α, ATF4 and CHOP, and promoted autophagy of osteoblasts via regulation of eIF2α, Atg7, LC3, and p62. Salubrinal markedly alleviated OVX-induced symptoms such as reduction of bone mineral density and bone volume fraction. In primary bone-marrow-derived cells, salubrinal increased the differentiation of osteoblasts, and decreased the formation of osteoclasts by inhibiting nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). Live cell imaging and RNA interference demonstrated that suppression of osteoclastogenesis is in part mediated by Rac1 GTPase. Collectively, this study demonstrates that ER stress-autophagy axis plays an important role in OVX mice. Bone-forming osteoblasts are restored by maintaining phosphorylation of eIF2α, and bone-resorbing osteoclasts are regulated by inhibiting NFATc1 and Rac1 GTPase.
Collapse
Affiliation(s)
- Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China
| | - Kazunori Hamamura
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Qiaoqiao Wan
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China.
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China.
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, 300052, Tianjin, China.
| |
Collapse
|
30
|
Baroncelli M, Drabek K, Eijken M, van der Eerden BCJ, van de Peppel J, van Leeuwen JPTM. Two-day-treatment of Activin-A leads to transient change in SV-HFO osteoblast gene expression and reduction in matrix mineralization. J Cell Physiol 2019; 235:4865-4877. [PMID: 31667867 PMCID: PMC7028110 DOI: 10.1002/jcp.29365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
Abstract
Activins regulate bone formation by controlling osteoclasts and osteoblasts. We investigated Activin‐A mechanism of action on human osteoblast mineralization, RNA and microRNA (miRNA) expression profile. A single 2‐day treatment of Activin‐A at Day 5 of osteoblast differentiation significantly reduced matrix mineralization. Activin A‐treated osteoblasts responded with transient change in gene expression, in a 2‐wave‐fashion. The 38 genes differentially regulated during the first wave (within 8 hr after Activin A start) were involved in transcription regulation. In the second wave (1–2 days after Activin A start), 65 genes were differentially regulated and related to extracellular matrix. Differentially expressed genes in both waves were associated to transforming growth factor beta signaling. We identified which microRNAs modulating osteoblast differentiation were regulated by Activin‐A. In summary, 2‐day treatment with Activin‐A in premineralization period of osteoblast cultures influenced miRNAs, gene transcription, and reduced matrix mineralization. Modulation of Activin A signaling might be useful to control bone quality for therapeutic purposes.
Collapse
Affiliation(s)
- Marta Baroncelli
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ksenija Drabek
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marco Eijken
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
31
|
4-Phenylbutyric Acid Reduces Endoplasmic Reticulum Stress in Chondrocytes That Is Caused by Loss of the Protein Disulfide Isomerase ERp57. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6404035. [PMID: 31781343 PMCID: PMC6875354 DOI: 10.1155/2019/6404035] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/01/2019] [Indexed: 01/30/2023]
Abstract
Objective The integrity of cartilage depends on the correct synthesis of extracellular matrix (ECM) components. In case of insufficient folding of proteins in the endoplasmic reticulum (ER) of chondrocytes, ECM proteins aggregate, ER stress evolves, and the unfolded protein response (UPR) is initiated. By this mechanism, chondrocytes relieve the stress condition or initiate cell death by apoptosis. Especially persistent ER stress has emerged as a pathogenic mechanism in cartilage diseases, such as chondrodysplasias and osteoarthritis. As pharmacological intervention is not available yet, it is of great interest to understand cartilage ER stress in detail and to develop therapeutics to intervene. Methods ERp57-deficient chondrocytes were generated by CRISPR/Cas9-induced KO. ER stress and autophagy were studied on mRNA and protein level as well as by transmission electron microscopy (TEM) in chondrocyte micromass or cartilage explant cultures of ERp57 KO mice. Thapsigargin (Tg), an inhibitor of the ER-residing Ca2+-ATPase, and 4-Phenylbutyric acid (4-PBA), a small molecular chemical chaperone, were applied to induce or inhibit ER stress. Results Our data reveal that the loss of the protein disulfide isomerase ERp57 is sufficient to induce ER stress in chondrocytes. 4-PBA efficiently diffuses into cartilage explant cultures and diminishes excessive ER stress in chondrocytes dose dependently, no matter if it is induced by ERp57 KO or stimulation with Tg. Conclusion ER-stress-related diseases have different sources; therefore, various targets for therapeutic treatment exist. In the future, 4-PBA may be used alone or in combination with other drugs for the treatment of ER-stress-related skeletal disorders in patients.
Collapse
|
32
|
Zheng W, Xie W, Yin D, Luo R, Liu M, Guo F. ATG5 and ATG7 induced autophagy interplays with UPR via PERK signaling. Cell Commun Signal 2019; 17:42. [PMID: 31060556 PMCID: PMC6503447 DOI: 10.1186/s12964-019-0353-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/13/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Autophagy and ER stress are involved in maintaining some well-orchestrated mechanisms aimed at either restoring cellular homeostasis or performing cell death. Autophagy is a well-defined process which governs overall cellular stress outcomes. Selective degradation of the ER mediated by autophagy occurs through a specific type of autophagy called ER-phagy, which ensures ER protein homeostasis. METHODS Immunoblotting and RT-PCR were used to evaluate the expression of ATG5 and ATG7 in chondrocyte. Western blotting, Flow cytometry,immunofluorescence cell staining and confocal microscope were used to examine the effect of ATG5 and ATG7 on autophagy, ER stress, cell apoptosis and cell proliferation. Transmission electron microscope and confocal microscope were performed to visualize the autophagy flux and autolysosome formation. The role of ATG5 and ATG7 overexpression on the PERK pathway inhibitor were detected by immunoblotting and treatment with inhibitors. RESULTS In current study, we demonstrated that Tm-induced ER stress can activate autophagy while Rapamycin-induced autophagy can inhibit ER stress in chondrocyte. Autophagy related protein ATG5 or ATG7 can promote autophagy and inhibit ER stress individually, and their combined effect can further improve the autophagy enhancement and the ER stress repression. Moreover, ATG5, ATG7 and ATG5 + ATG7 lead cells into more S phase, increase the number of S phase and inhibit apoptosis as well. ATG5, ATG7 and ATG5 + ATG7 regulate autophagy, ER stress, apoptosis and cell cycle through PERK signaling, a vital UPR branch pathway. CONCLUSIONS ATG5 and ATG7 connect autophagy with ER stress through PERK signaling. The protective effect of ATG5/7 overexpression on chondrocyte survival relys on PERK signaling. The effect of siPERK and siNrf2 on the cytoprotective effect of ATG5/7 are of synergism, while the effect of siPERK and siATF4 are of antagonism. PERK signal may be the pivot for autophagy, ER homeostasis and ER-phagy in chondrocyte.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016 China
| | - Weiwei Xie
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016 China
| | - Danyang Yin
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016 China
| | - Rui Luo
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016 China
| | - Min Liu
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016 China
| | - Fengjin Guo
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
33
|
Zheng C, Lin X, Xu X, Wang C, Zhou J, Gao B, Fan J, Lu W, Hu Y, Jie Q, Luo Z, Yang L. Suppressing UPR-dependent overactivation of FGFR3 signaling ameliorates SLC26A2-deficient chondrodysplasias. EBioMedicine 2019; 40:695-709. [PMID: 30685387 PMCID: PMC6413327 DOI: 10.1016/j.ebiom.2019.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Background Mutations in the SLC26A2 gene cause a spectrum of currently incurable human chondrodysplasias. However, genotype-phenotype relationships of SLC26A2-deficient chondrodysplasias are still perplexing and thus stunt therapeutic development. Methods To investigate the causative role of SLC26A2 deficiency in chondrodysplasias and confirm its skeleton-specific pathology, we generated and analyzed slc26a2−/− and Col2a1-Cre; slc26a2fl/fl mice. The therapeutic effect of NVP-BGJ398, an FGFR inhibitor, was tested with both explant cultures and timed pregnant females. Findings Two lethal forms of human SLC26A2-related chondrodysplasias, achondrogenesis type IB (ACG1B) and atelosteogenesis type II (AO2), are phenocopied by slc26a2−/− mice. Unexpectedly, slc26a2−/− chondrocytes are defective for collagen secretion, exhibiting intracellular retention and compromised extracellular deposition of ColII and ColIX. As a consequence, the ATF6 arm of the unfolded protein response (UPR) is preferentially triggered to overactivate FGFR3 signaling by inducing excessive FGFR3 in slc26a2−/− chondrocytes. Consistently, suppressing FGFR3 signaling by blocking either FGFR3 or phosphorylation of the downstream effector favors the recovery of slc26a2−/− cartilage cultures from impaired growth and unbalanced cell proliferation and apoptosis. Moreover, administration of an FGFR inhibitor to pregnant females shows therapeutic effects on pathological features in slc26a2−/− newborns. Finally, we confirm the skeleton-specific lethality and pathology of global SLC26A2 deletion through analyzing the Col2a1-Cre; slc26a2fl/fl mouse line. Interpretation Our study unveils a previously unrecognized pathogenic mechanism underlying ACG1B and AO2, and supports suppression of FGFR3 signaling as a promising therapeutic approach for SLC26A2-related chondrodysplasias. Fund This work was supported by National Natural Science Foundation of China (81871743, 81730065 and 81772377).
Collapse
Affiliation(s)
- Chao Zheng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xisheng Lin
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaolong Xu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Cheng Wang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Jinru Zhou
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Bo Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Fan
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weiguang Lu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yaqian Hu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qiang Jie
- Department of Orthopedic Surgery, HongHui Hospital, Xi'an Jiaotong University, College of Medicine, Xi'an, China
| | - Zhuojing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Medical Research Institute, Northwestern Polytechnical University, Xi'an, China
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China; Medical Research Institute, Northwestern Polytechnical University, Xi'an, China.
| |
Collapse
|
34
|
Different Forms of ER Stress in Chondrocytes Result in Short Stature Disorders and Degenerative Cartilage Diseases: New Insights by Cartilage-Specific ERp57 Knockout Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8421394. [PMID: 30647818 PMCID: PMC6311764 DOI: 10.1155/2018/8421394] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
Abstract
Cartilage is essential for skeletal development by endochondral ossification. The only cell type within the tissue, the chondrocyte, is responsible for the production of macromolecules for the extracellular matrix (ECM). Before proteins and proteoglycans are secreted, they undergo posttranslational modification and folding in the endoplasmic reticulum (ER). However, the ER folding capacity in the chondrocytes has to be balanced with physiological parameters like energy and oxygen levels. Specific cellular conditions, e.g., a high protein demand, or pathologic situations disrupt ER homeostasis and lead to the accumulation of poorly folded or misfolded proteins. This state is called ER stress and induces a cellular quality control system, the unfolded protein response (UPR), to restore homeostasis. Different mouse models with ER stress in chondrocytes display comparable skeletal phenotypes representing chondrodysplasias. Therefore, ER stress itself seems to be involved in the pathogenesis of these diseases. It is remarkable that chondrodysplasias with a comparable phenotype arise independent from the sources of ER stress, which are as follows: (1) mutations in ECM proteins leading to aggregation, (2) deficiencies in ER chaperones, (3) mutations in UPR signaling factors, or (4) deficiencies in the degradation of aggregated proteins. In any case, the resulting UPR substantially impairs ECM protein synthesis, chondrocyte proliferation, and/or differentiation or regulation of autophagy and apoptosis. Notably, chondrodysplasias arise no matter if single or multiple events are affected. We analyzed cartilage-specific ERp57 knockout mice and demonstrated that the deficiency of this single protein disulfide isomerase, which is responsible for formation of disulfide bridges in ECM glycoproteins, is sufficient to induce ER stress and to cause an ER stress-related bone phenotype. These mice therefore qualify as a novel model for the analysis of ER stress in chondrocytes. They give new insights in ER stress-related short stature disorders and enable the analysis of ER stress in other cartilage diseases, such as osteoarthritis.
Collapse
|
35
|
Doron B, Abdelhamed S, Butler JT, Hashmi SK, Horton TM, Kurre P. Transmissible ER stress reconfigures the AML bone marrow compartment. Leukemia 2018; 33:918-930. [PMID: 30206307 PMCID: PMC6411460 DOI: 10.1038/s41375-018-0254-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022]
Abstract
Successive adaptation of the bone marrow (BM) from homeostatic hematopoietic microenvironment to a self-reinforcing niche is an integral aspect of leukemogenesis. Yet, the cellular mechanisms underlying these functional alterations remain to be defined. Here, we found that AML incursion precipitates compartmental endoplasmic reticulum (ER) stress and an unfolded protein response (UPR) in both leukemia and stromal cells. We observed that extracellular vesicles (EV) transmit ER stress in vivo from the AML xenograft to BM stroma, whereby the upregulation of core UPR components drives subsequent osteolineage differentiation of mesenchymal stem cells (MSC). Finally, we show that the underlying mechanism involves quantitative incorporation and cell-cell transfer of Bone Morphogenic Protein 2 (BMP2), a potent osteogenic signal, by AML-EVs. Corroborative studies in AML patient samples support the translational relevance of AML-EVs as a platform for BMP trafficking and source of compartmental crosstalk. Transmissible ER stress was previously identified as a source of chemoresistance in solid tumor models, and this work reveals a role in remodeling the BM niche in AML.
Collapse
Affiliation(s)
- Ben Doron
- Department of Pediatrics, Pediatric Blood & Cancer Biology Program, Papé Family Pediatric Research Institute, Portland, OR, USA.,OHSU Knight Cancer Institute, Portland, OR, USA.,Department of Biomedical Engineering, Portland, OR, USA
| | - Sherif Abdelhamed
- Department of Pediatrics, Pediatric Blood & Cancer Biology Program, Papé Family Pediatric Research Institute, Portland, OR, USA.,OHSU Knight Cancer Institute, Portland, OR, USA.,Department of Biomedical Engineering, Portland, OR, USA
| | - John T Butler
- Department of Pediatrics, Pediatric Blood & Cancer Biology Program, Papé Family Pediatric Research Institute, Portland, OR, USA.,OHSU Knight Cancer Institute, Portland, OR, USA.,Department of Biomedical Engineering, Portland, OR, USA.,Oregon Health & Science University, Portland, OR, USA
| | - Saman K Hashmi
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX, USA
| | - Terzah M Horton
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX, USA
| | - Peter Kurre
- Department of Pediatrics, Pediatric Blood & Cancer Biology Program, Papé Family Pediatric Research Institute, Portland, OR, USA. .,OHSU Knight Cancer Institute, Portland, OR, USA. .,Department of Biomedical Engineering, Portland, OR, USA. .,Division of Hematology, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, ARC 302, Philadelphia, 19104, PA, USA.
| |
Collapse
|
36
|
Wang C, Tan Z, Niu B, Tsang KY, Tai A, Chan WCW, Lo RLK, Leung KKH, Dung NWF, Itoh N, Zhang MQ, Chan D, Cheah KSE. Inhibiting the integrated stress response pathway prevents aberrant chondrocyte differentiation thereby alleviating chondrodysplasia. eLife 2018; 7:37673. [PMID: 30024379 PMCID: PMC6053305 DOI: 10.7554/elife.37673] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/05/2018] [Indexed: 12/16/2022] Open
Abstract
The integrated stress response (ISR) is activated by diverse forms of cellular stress, including endoplasmic reticulum (ER) stress, and is associated with diseases. However, the molecular mechanism(s) whereby the ISR impacts on differentiation is incompletely understood. Here, we exploited a mouse model of Metaphyseal Chondrodysplasia type Schmid (MCDS) to provide insight into the impact of the ISR on cell fate. We show the protein kinase RNA-like ER kinase (PERK) pathway that mediates preferential synthesis of ATF4 and CHOP, dominates in causing dysplasia by reverting chondrocyte differentiation via ATF4-directed transactivation of Sox9. Chondrocyte survival is enabled, cell autonomously, by CHOP and dual CHOP-ATF4 transactivation of Fgf21. Treatment of mutant mice with a chemical inhibitor of PERK signaling prevents the differentiation defects and ameliorates chondrodysplasia. By preventing aberrant differentiation, titrated inhibition of the ISR emerges as a rationale therapeutic strategy for stress-induced skeletal disorders.
Collapse
Affiliation(s)
- Cheng Wang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Zhijia Tan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Ben Niu
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Kwok Yeung Tsang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Andrew Tai
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Wilson C W Chan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Rebecca L K Lo
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Keith K H Leung
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Nelson W F Dung
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, University of Kyoto, Kyoto, Japan
| | - Michael Q Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Richardson, United States.,MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Danny Chan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
37
|
Fujii T, Fujita N, Suzuki S, Tsuji T, Takaki T, Umezawa K, Watanabe K, Miyamoto T, Horiuchi K, Matsumoto M, Nakamura M. The unfolded protein response mediated by PERK is casually related to the pathogenesis of intervertebral disc degeneration. J Orthop Res 2018; 36:1334-1345. [PMID: 29080374 DOI: 10.1002/jor.23787] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/26/2017] [Indexed: 02/04/2023]
Abstract
Although the number of patients with intervertebral disc (IVD) degeneration is increasing in aging societies, its etiology and pathogenesis remain elusive and there is currently no effective treatment to prevent this undesirable condition. The unfolded protein response (UPR) is a cellular machinery that plays critical roles in handling endoplasmic reticulum (ER) stress, a condition caused by the accumulation of unfolded proteins in the ER lumen. This study aimed to elucidate the potential role of the UPR mediated by pancreatic endoplasmic reticulum kinase (PERK), one of the major ER stress sensors in mammalian cells, in the development of IVD degeneration. IVD degeneration was artificially induced in Wister rats by percutaneously puncturing the coccyx IVDs and human IVDs were collected from patients who underwent spinal surgery. Expression of the UPR target genes was elevated in degenerative IVDs in both humans and rats. The induction of ER stress in annulus fibrosus cells significantly increased the transcripts for tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in a nuclear factor (NF)-κB pathway-dependent manner. The expression of TNF-α and IL-6 was significantly reduced by treatment with a selective PERK inhibitor, GSK2606414, and by gene silencing against PERK and activating transcription factor 4 (ATF4) transcripts. Our findings indicate that the UPR mediated by the PERK pathway is causally related to the development of IVD degeneration, suggesting that PERK may be a potential molecular target for suppressing the degenerative changes in IVDs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1334-1345, 2018.
Collapse
Affiliation(s)
- Takeshi Fujii
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Nobuyuki Fujita
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Satoshi Suzuki
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Takashi Tsuji
- Department of Orthopaedic Surgery, Fujita Health University, Aichi, Japan
| | - Takashi Takaki
- Section of Electron Microscopy, Showa University, Tokyo, Japan.,Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuo Umezawa
- Department of Molecular Target Medicine, Aichi Medical University School of Medicine, Aichi, Japan
| | - Kota Watanabe
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Takeshi Miyamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Keisuke Horiuchi
- Department of Orthopaedic Surgery, National Defence Medical College, Saitama, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
38
|
Yang D, Jiang T, Liu J, Hong J, Lin P, Chen H, Zhou D, Tang K, Wang A, Jin Y. Hormone regulates endometrial function via cooperation of endoplasmic reticulum stress and mTOR-autophagy. J Cell Physiol 2018; 233:6644-6659. [PMID: 29206294 DOI: 10.1002/jcp.26315] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/03/2017] [Indexed: 12/18/2022]
Abstract
In ruminant, the receptive endometrium and the elongation of the hatched blastocyst are required to complete the process of implantation. However, the mechanisms regulating goat endometrial function during the peri-implantation period of pregnancy are still unclear. In this study, EECs were treated with progesterone, estradiol, and interferon-tau (IFNT). We have found that endoplasmic reticulum (ER) stress was activated under hormones treatment. To identify the cellular mechanism of regulation of endometrial function, we investigated the effect of ER stress activator thapsigargin (TG) and inhibitor 4 phenyl butyric acid (4-PBA) on EECs. We found that TG, which activated the three branches of UPR, increased the expression of genes associated with promoting conceptus elongation and cellular attachment, significantly up-regulated the spheroid attachment rate and PGE2 /PGF2α ratio. 4-PBA pre-treatment inhibited UPR and inhibited promoting conceptus elongation and cellular attachment related genes, but the spheroid attachment rate and PGE2 /PGF2α ratio were not changed significantly. Moreover, knockdown of ATF6 via shATF6 promoted the conceptus elongation related genes, but increased the dissolution of the corpus luteum. Besides, blocking ATF6 attenuated autophagy by activating mammalian target of rapamycin (mTOR) pathway. Moreover, rapamycin (mTOR inhibitor) pre-treatment inhibited the expression of promoting conceptus elongation and increased PGE2 /PGF2α ratio. Taken together, our study indicated that physiological level of ER stress may contribute to early pregnancy success, and ATF6 signaling pathway cooperated with autophagy to regulate endometrial function by modulating mTOR pathway.
Collapse
Affiliation(s)
- Diqi Yang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Tingting Jiang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianguo Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jin Hong
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Huatao Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Dong Zhou
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Keqiong Tang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
39
|
PERK-mediated translational control is required for collagen secretion in chondrocytes. Sci Rep 2018; 8:773. [PMID: 29335505 PMCID: PMC5768675 DOI: 10.1038/s41598-017-19052-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/20/2017] [Indexed: 12/22/2022] Open
Abstract
As chondrocytes are highly secretory and they experience a variety of stresses, physiological unfolded protein response (UPR) signalling is essential for extracellular matrix (ECM) secretion and chondrogenesis. In the three branches of the UPR pathway, PERK governs the translational attenuation and transcriptional upregulation of amino acid and redox metabolism and induction of apoptosis. It was previously demonstrated that a defect of the PERK branch of the UPR signalling pathway causes the accumulation of unfolded proteins, leading to cell death without perturbing endoplasmic reticulum (ER)-to-Golgi transport in pancreatic β cells. However, little is known about the role of PERK in chondrocytes. In this study, we found that PERK signalling is activated in chondrocytes, and inhibition of PERK reduces collagen secretion despite causing excessive collagen synthesis in the ER. Perk−/− mice displayed reduced collagen in articular cartilage but no differences in chondrocyte proliferation or apoptosis compared to the findings in wild-type mice. PERK inhibition increases misfolded protein levels in the ER, which largely hinder ER-to-Golgi transport. These results suggest that the translational control mediated by PERK is a critical determinant of ECM secretion in chondrocytes.
Collapse
|
40
|
Webb EA, Balasubramanian M, Fratzl-Zelman N, Cabral WA, Titheradge H, Alsaedi A, Saraff V, Vogt J, Cole T, Stewart S, Crabtree NJ, Sargent BM, Gamsjaeger S, Paschalis EP, Roschger P, Klaushofer K, Shaw NJ, Marini JC, Högler W. Phenotypic Spectrum in Osteogenesis Imperfecta Due to Mutations in TMEM38B: Unraveling a Complex Cellular Defect. J Clin Endocrinol Metab 2017; 102:2019-2028. [PMID: 28323974 PMCID: PMC5470761 DOI: 10.1210/jc.2016-3766] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/09/2017] [Indexed: 12/19/2022]
Abstract
CONTEXT Recessive mutations in TMEM38B cause type XIV osteogenesis imperfecta (OI) by dysregulating intracellular calcium flux. OBJECTIVES Clinical and bone material phenotype description and osteoblast differentiation studies. DESIGN AND SETTING Natural history study in pediatric research centers. PATIENTS Eight patients with type XIV OI. MAIN OUTCOME MEASURES Clinical examinations included bone mineral density, radiographs, echocardiography, and muscle biopsy. Bone biopsy samples (n = 3) were analyzed using histomorphometry, quantitative backscattered electron microscopy, and Raman microspectroscopy. Cellular differentiation studies were performed on proband and control osteoblasts and normal murine osteoclasts. RESULTS Type XIV OI clinical phenotype ranges from asymptomatic to severe. Previously unreported features include vertebral fractures, periosteal cloaking, coxa vara, and extraskeletal features (muscular hypotonia, cardiac abnormalities). Proband lumbar spine bone density z score was reduced [median -3.3 (range -4.77 to +0.1; n = 7)] and increased by +1.7 (1.17 to 3.0; n = 3) following bisphosphonate therapy. TMEM38B mutant bone has reduced trabecular bone volume, osteoblast, and particularly osteoclast numbers, with >80% reduction in bone resorption. Bone matrix mineralization is normal and nanoporosity low. We demonstrate a complex osteoblast differentiation defect with decreased expression of early markers and increased expression of late and mineralization-related markers. Predominance of trimeric intracellular cation channel type B over type A expression in murine osteoclasts supports an intrinsic osteoclast defect underlying low bone turnover. CONCLUSIONS OI type XIV has a bone histology, matrix mineralization, and osteoblast differentiation pattern that is distinct from OI with collagen defects. Probands are responsive to bisphosphonates and some show muscular and cardiovascular features possibly related to intracellular calcium flux abnormalities.
Collapse
Affiliation(s)
- Emma A. Webb
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham B4 6NH, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
| | - Meena Balasubramanian
- Sheffield Clinical Genetics Service, Sheffield Children’s National Health Service Foundation Trust, Sheffield S10 2TH United Kingdom
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt Trama Centre Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Wayne A. Cabral
- Section on Heritable Disorders of Bone and Extracellular Matrix, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Hannah Titheradge
- Department of Clinical Genetics, Birmingham Women’s Hospital, Birmingham B15 2DG, United Kingdom
| | - Atif Alsaedi
- Department of Clinical Genetics, Birmingham Women’s Hospital, Birmingham B15 2DG, United Kingdom
| | - Vrinda Saraff
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham B4 6NH, United Kingdom
| | - Julie Vogt
- Department of Clinical Genetics, Birmingham Women’s Hospital, Birmingham B15 2DG, United Kingdom
| | - Trevor Cole
- Department of Clinical Genetics, Birmingham Women’s Hospital, Birmingham B15 2DG, United Kingdom
| | - Susan Stewart
- Department of Clinical Genetics, Birmingham Women’s Hospital, Birmingham B15 2DG, United Kingdom
| | - Nicola J. Crabtree
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham B4 6NH, United Kingdom
| | - Brandi M. Sargent
- Section on Heritable Disorders of Bone and Extracellular Matrix, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Sonja Gamsjaeger
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt Trama Centre Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Eleftherios P. Paschalis
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt Trama Centre Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Paul Roschger
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt Trama Centre Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Klaus Klaushofer
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of Wiener Gebietskrankenkasse and Allgemeine Unfallversicherungsanstalt Trama Centre Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Nick J. Shaw
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham B4 6NH, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
| | - Joan C. Marini
- Section on Heritable Disorders of Bone and Extracellular Matrix, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Wolfgang Högler
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham B4 6NH, United Kingdom
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
| |
Collapse
|
41
|
Wu Z, Li M, Zheng W, Hu Q, Cheng Z, Guo F. Silencing of both ATF4 and PERK inhibits cell cycle progression and promotes the apoptosis of differentiating chondrocytes. Int J Mol Med 2017; 40:101-111. [PMID: 28498443 PMCID: PMC5466379 DOI: 10.3892/ijmm.2017.2985] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/25/2017] [Indexed: 01/06/2023] Open
Abstract
In the current study, we demonstrate that the silencing of protein kinase R (PKR)-like endoplasmic reticulum (ER) kinase (PERK) and activating transcription factor 6 (ATF4) (using small interfering RNA expression constructs) inhibits the chondrocyte cell cycle and proliferation in vitro and ex vivo. The silencing of PERK alone using siRNA against PERK (siPERK) led to arrest in the G1 phase, it decreased the number of cells in the S phase, and delayed progressoin to the G2-M phase. Co-transfection with siRNA against ATF (siATF4) led to a more profound inhibitory effect on cell cycle progression. Moreover, transfection with siPERK was associated with enhanced endoplasmic reticulum (ER) stress-induced apoptosis during bone morphogenetic protein 2 (BMP2)-induced chondrogenesis, and transfection with siATF4 exacerbated ER stress-related cell death. Data from flow cytometry (FCM), immunohistochemistry and TUNEL assays supported these findings in vitro and ex vivo. As shown by our results, the combined effect of the silencing of ATF4 and PERK led to the activation of an ER stress-specific caspase cascade in the cartilage tissue. On the whole, these findings reveal a new crucial combined effect of the silencing of PERK and ATF4 in modulating ER stress-mediated apoptosis during chondrocyte differentiation and proliferation.
Collapse
Affiliation(s)
- Zhimeng Wu
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Meiling Li
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wei Zheng
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qin Hu
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhi Cheng
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Fengjin Guo
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
42
|
Liu CF, Samsa WE, Zhou G, Lefebvre V. Transcriptional control of chondrocyte specification and differentiation. Semin Cell Dev Biol 2016; 62:34-49. [PMID: 27771362 DOI: 10.1016/j.semcdb.2016.10.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022]
Abstract
A milestone in the evolutionary emergence of vertebrates was the invention of cartilage, a tissue that has key roles in modeling, protecting and complementing the bony skeleton. Cartilage is elaborated and maintained by chondrocytes. These cells derive from multipotent skeletal progenitors and they perform highly specialized functions as they proceed through sequential lineage commitment and differentiation steps. They form cartilage primordia, the primary skeleton of the embryo. They then transform these primordia either into cartilage growth plates, temporary drivers of skeletal elongation and endochondral ossification, or into permanent tissues, namely articular cartilage. Chondrocyte fate decisions and differentiated activities are controlled by numerous extrinsic and intrinsic cues, and they are implemented at the gene expression level by transcription factors. The latter are the focus of this review. Meritorious efforts from many research groups have led over the last two decades to the identification of dozens of key chondrogenic transcription factors. These regulators belong to all types of transcription factor families. Some have master roles at one or several differentiation steps. They include SOX9 and RUNX2/3. Others decisively assist or antagonize the activities of these masters. They include TWIST1, SOX5/6, and MEF2C/D. Many more have tissue-patterning roles and regulate cell survival, proliferation and the pace of cell differentiation. They include, but are not limited to, homeodomain-containing proteins and growth factor signaling mediators. We here review current knowledge of all these factors, one superclass, class, and family at a time. We then compile all knowledge into transcriptional networks. We also identify remaining gaps in knowledge and directions for future research to fill these gaps and thereby provide novel insights into cartilage disease mechanisms and treatment options.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA.
| | - William E Samsa
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA
| | - Guang Zhou
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Véronique Lefebvre
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA.
| |
Collapse
|