1
|
Cillo M, Buonomo V, Vainshtein A, Grumati P. Autophagy, ER-phagy and ER dynamics during cell differentiation. J Mol Biol 2025:169151. [PMID: 40222412 DOI: 10.1016/j.jmb.2025.169151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle essential for protein and lipid synthesis, ion transport and inter-organelle communication. It comprises a highly dynamic network of membranes that continuously reshape to support a wide range of cellular processes. During cellular differentiation, extensive remodelling of both ER architecture and its proteome is required to accommodate alterations in cell morphology and function. Autophagy, and ER-phagy in particular, plays a pivotal role in reshaping the ER, enabling cells to meet their evolving needs and adapt to developmental cues. Despite the ER's critical role in cellular differentiation, the mechanisms responsible for regulating its dynamics are not fully understood. Emerging evidence suggests that transcriptional and post-translational regulation play a role in fine-tuning ER-phagy and the unfolded protein response (UPR). This review explores the molecular basis of autophagy and ER-phagy, highlighting their role in ER remodelling during cellular differentiation. A deeper understanding of these processes could open new avenues for targeted therapeutic approaches in conditions where ER remodelling is impaired.
Collapse
Affiliation(s)
- Michele Cillo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Viviana Buonomo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | | | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy.
| |
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
|
Haacke N, Wang H, Yan S, Barovic M, Li X, Nagai K, Botezatu A, Hatzioannou A, Gercken B, Trimaglio G, Shah AU, Wang J, Ye L, Jaykar MT, Rauner M, Wielockx B, Chung KJ, Netea MG, Kalafati L, Hajishengallis G, Chavakis T. Innate immune training of osteoclastogenesis promotes inflammatory bone loss in mice. Dev Cell 2025:S1534-5807(25)00063-2. [PMID: 40020679 PMCID: PMC7617534 DOI: 10.1016/j.devcel.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
We previously demonstrated that long-term trained immunity (TRIM) involves adaptations that imprint innate immune memory in long-lived myelopoiesis precursors and their progeny, monocytes/macrophages and neutrophils, which thereby acquire enhanced responsiveness to future challenges. Here, we show that a distinct component of myeloid biology, osteoclastogenesis, can also undergo innate immune training. Indeed, β-glucan-induced TRIM was associated with an increased osteoclastogenesis bias in the bone marrow and an expansion of monocytes/osteoclast progenitors in the periphery, resulting in aggravated severity of experimental periodontitis and arthritis. In the setting of trained inflammatory osteoclastogenesis, we observed transcriptomic rewiring in synovial myeloid cells of arthritic mice, featuring prominent upregulation of the transcription factor melanogenesis-associated transcription factor (MITF). Adoptive transfer of splenic monocytes from β-glucan-trained mice to naive recipients exacerbated arthritis in the latter in a strictly MITF-dependent manner. Our findings establish trained osteoclastogenesis as a maladaptive component of TRIM and potentially provide therapeutic targets in inflammatory bone loss disorders.
Collapse
Affiliation(s)
- Nora Haacke
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shu Yan
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Marko Barovic
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kosuke Nagai
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Adelina Botezatu
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Aikaterini Hatzioannou
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Giulia Trimaglio
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany
| | - Anisha U Shah
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mangesh T Jaykar
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 XZ Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Lydia Kalafati
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; National Center for Tumor Diseases, Partner Site Dresden, 01307 Dresden, Germany; Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| |
Collapse
|
4
|
Yang Z, Hou N, Cheng W, Lu X, Wang M, Bai S, Lin Y, Wang Y, Lin S, Zhang P, Tortorella MD, Feng L, Li G. MiR-378 exaggerates angiogenesis and bone erosion in collagen-induced arthritis mice by regulating endoplasmic reticulum stress. Cell Death Dis 2024; 15:910. [PMID: 39695085 DOI: 10.1038/s41419-024-07193-5] [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: 05/07/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 12/20/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder marked by pain, inflammation, and discomfort in the synovial joints. It is critical to understand the pathological mechanisms of RA progression. MicroRNA-378 (miR-378) is highly expressed in the synovium of RA patients and positively correlated with disease severity, but its function and underlying mechanisms remain poorly understood. In this study, miR-378 transgenic (miR-378high) mice were used to construct the collagen-induced arthritis (CIA) model for exploring the role of miR-378 in RA development. miR-378high CIA mice showed accelerated RA development, as evidenced by exaggerated joint swelling and bone structural deformities. More severe endoplasmic reticulum (ER) stress and the consequent angiogenesis and osteoclastogenesis were also activated in the synovial tissue and calcaneus, respectively, in the miR-378high group, suggesting that ER plays a significant role in miR-378-mediated RA pathogenesis. Upon in vitro RA induction, fibroblast-like synoviocytes (FLSs) isolated from miR-378high mice showed a higher expression level of ER stress markers. The conditioned medium (CM) from RA-FLSs of miR-378high mice stimulated more intensive angiogenesis and osteoclastogenesis. The ER stress-related protein Crebrf was identified as a downstream target of miR-378. Crebrf knockdown diminished the promoting effect of miR-378 on ER stress, as well as its downstream angiogenesis and osteoclastogenesis activities. Tail vein injection of anti-miR-378 lentivirus in an established RA mouse model was shown to ameliorate RA progression. In conclusion, miR-378 amplified RA development by promoting ER stress and downstream angiogenesis and osteoclastogenesis, thus indicating that miR-378 may be a potential therapeutic target for RA treatment.
Collapse
Grants
- 82172430 National Science Foundation of China | National Natural Science Foundation of China-Yunnan Joint Fund (NSFC-Yunnan Joint Fund)
- 82272505 National Science Foundation of China | National Natural Science Foundation of China-Yunnan Joint Fund (NSFC-Yunnan Joint Fund)
- 14108720 Research Grants Council, University Grants Committee (RGC, UGC)
- 14121721 Research Grants Council, University Grants Committee (RGC, UGC)
- 14202920 Research Grants Council, University Grants Committee (RGC, UGC)
- N_CUHK472/22 Research Grants Council, University Grants Committee (RGC, UGC)
- C7030-18G Research Grants Council, University Grants Committee (RGC, UGC)
- T13-402/17-N Research Grants Council, University Grants Committee (RGC, UGC)
- AoE/M-402/20 Research Grants Council, University Grants Committee (RGC, UGC)
- 17180831 Food and Health Bureau of the Government of the Hong Kong Special Administrative Region | Health and Medical Research Fund (HMRF)
- 08190416 Food and Health Bureau of the Government of the Hong Kong Special Administrative Region | Health and Medical Research Fund (HMRF)
- 09203436 Food and Health Bureau of the Government of the Hong Kong Special Administrative Region | Health and Medical Research Fund (HMRF)
- PRP/050/19FX Innovation and Technology Commission (ITF)
- Health@InnoHK Innovation and Technology Commission (ITF)
Collapse
Affiliation(s)
- Zhengmeng Yang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Nan Hou
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Wenxiang Cheng
- Institute of Translation and Medical Research and Development Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Xuan Lu
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Ming Wang
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Shanshan Bai
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, PR China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Yuejun Lin
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, PR China
| | - Sien Lin
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China
| | - Peng Zhang
- Institute of Translation and Medical Research and Development Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Micky D Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, PR China.
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, PR China.
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, PR China.
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, PR China.
| |
Collapse
|
5
|
Meng Q, Han J, Zhang X, Su W, Liu B, Liu T. Comprehensive Analysis of Immune Infiltration and Key Genes in Peri-Implantitis Using Bioinformatics and Molecular Biology Approaches. Med Sci Monit 2024; 30:e941870. [PMID: 39501535 PMCID: PMC11552188 DOI: 10.12659/msm.941870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/10/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND Peri-implantitis is the main cause of failure of implant treatment, and there is little research on its molecular mechanism. This study aimed to identify key biomarkers and immune infiltration of peri-implantitis using a bioinformatics method. MATERIAL AND METHODS Three Gene Ontology (GO) gene expression profiles were selected from the Gene Expression Omnibus. Differentially expressed genes (DEGs) were identified by the LIMMA package, and functional correlations of DEGs were analyzed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis. Information on immune-related genes was obtained from ImmPort (https://www.immport.org) and InnateDB (http://www.innatedb.com). Immune-related DEGs were screened by least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE). The single-sample Gene Set Enrichment Analysis algorithm was used to analyze immune cell infiltration in gingival tissue between peri-implantitis and normal controls. Finally, results of bioinformatics analysis were verified by qPCR. RESULTS A total of 398 DEGs were identified, of which 96 were immune-related. Enrichment analysis showed these genes were enriched in inflammatory response, leucocyte chemotaxis, immune response-regulating signaling pathway, and cell activation. Seven key genes were selected by LASSO and SVM-RFE. Receiver operating characteristic curve results showed these genes had excellent diagnostic efficacy. Results of qPCR showed significant differences in the expression of these genes. CONCLUSIONS Differences in key genes and immune infiltration between peri-implantitis and gingival tissues of normal controls may provide new insights into the development of peri-implantitis. Elucidating the difference in immune infiltration between peri-implantitis tissues and normal tissues will help to understand the development of peri-implantitis.
Collapse
Affiliation(s)
- Qingxun Meng
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, PR China
- School of Medicine, Nankai University, Tianjin, PR China
| | - Jing Han
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, PR China
- School of Medicine, Nankai University, Tianjin, PR China
| | - Xi Zhang
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, PR China
- School of Medicine, Nankai University, Tianjin, PR China
| | - Wenxuan Su
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, PR China
- School of Medicine, Nankai University, Tianjin, PR China
| | - Beibei Liu
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, PR China
- School of Medicine, Nankai University, Tianjin, PR China
| | - Taicheng Liu
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, PR China
- School of Medicine, Nankai University, Tianjin, PR China
| |
Collapse
|
6
|
Zhang Z, Zhang X, Wei X, Yu C, Xiao L, Liu J, Liu Y, Cao Y, Song K. IRE1α inhibits osteogenic differentiation of mouse embryonic fibroblasts by limiting Shh signaling. Oral Dis 2024; 30:4504-4517. [PMID: 38438324 DOI: 10.1111/odi.14919] [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: 09/25/2023] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024]
Abstract
OBJECTIVES This study aimed to investigate the effect of endoplasmic reticulum (ER) stress sensor inositol-requiring enzyme 1α (IRE1α) on the sonic hedgehog N-terminus (N-Shh)-enhanced-osteogenic differentiation process in mouse embryonic fibroblasts (MEFs). MATERIALS AND METHODS Osteogenesis of MEFs was observed by alkaline phosphatase (ALP) staining, alizarin red staining, and Von Kossa staining assays. Activation of unfolded protein response and Shh signaling were examined using real-time quantitative PCR and western blot assays. IRE1α-deficient MEFs were used to explore the effect of IRE1α on N-Shh-driven osteogenesis. RESULTS N-Shh increased ALP activity, matrix mineralization, and the expression of Alp and Col-I in MEFs under osteogenic conditions; notably, this was reversed when combined with the ER stress activator Tm treatment. Interestingly, the administration of N-Shh decreased the expression of IRE1α. Abrogation of IRE1α increased the expression of Shh pathway factors in osteogenesis-induced MEFs, contributing to the osteogenic effect of N-Shh. Moreover, IRE1α-deficient MEFs exhibited elevated levels of osteogenic markers. CONCLUSIONS Our findings suggest that the IRE1α-mediated unfolded protein response may alleviate the ossification of MEFs by attenuating Shh signaling. Our research has identified a strategy to inhibit excessive ossification, which may have clinical significance in preventing temporomandibular joint bony ankylosis.
Collapse
Affiliation(s)
- Zhixiang Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, Hubei, China
| | - Xuan Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, and the Institute for Advanced Studies, Wuhan University, Wuhan, Hubei, China
| | - Xiangzhen Wei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, and the Institute for Advanced Studies, Wuhan University, Wuhan, Hubei, China
| | - Chengbo Yu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, Hubei, China
| | - Li Xiao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, Hubei, China
| | - Jianmiao Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, and the Institute for Advanced Studies, Wuhan University, Wuhan, Hubei, China
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, Hubei, China
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, Hubei, China
| |
Collapse
|
7
|
Yu C, Zhang Z, Xiao L, Ai M, Qing Y, Zhang Z, Xu L, Yu OY, Cao Y, Liu Y, Song K. IRE1α pathway: A potential bone metabolism mediator. Cell Prolif 2024; 57:e13654. [PMID: 38736291 PMCID: PMC11471397 DOI: 10.1111/cpr.13654] [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: 12/28/2023] [Revised: 04/07/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Osteoblasts and osteoclasts collaborate in bone metabolism, facilitating bone development, maintaining normal bone density and strength, and aiding in the repair of pathological damage. Endoplasmic reticulum stress (ERS) can disrupt the intracellular equilibrium between osteoclast and osteoblast, resulting in dysfunctional bone metabolism. The inositol-requiring enzyme-1α (IRE1α) pathway-the most conservative unfolded protein response pathway activated by ERS-is crucial in regulating cell metabolism. This involvement encompasses functions such as inflammation, autophagy, and apoptosis. Many studies have highlighted the potential roles of the IRE1α pathway in osteoblasts, chondrocytes, and osteoclasts and its implication in certain bone-related diseases. These findings suggest that it may serve as a mediator for bone metabolism. However, relevant reviews on the role of the IRE1α pathway in bone metabolism remain unavailable. Therefore, this review aims to explore recent research that elucidated the intricate roles of the IRE1α pathway in bone metabolism, specifically in osteogenesis, chondrogenesis, osteoclastogenesis, and osteo-immunology. The findings may provide novel insights into regulating bone metabolism and treating bone-related diseases.
Collapse
Affiliation(s)
- Chengbo Yu
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Zhixiang Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Li Xiao
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Mi Ai
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Ying Qing
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Zhixing Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Lianyi Xu
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Ollie Yiru Yu
- Faculty of DentistryThe University of Hong KongHong Kong SARChina
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, and the Institute for Advanced StudiesWuhan UniversityWuhanHubeiChina
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| |
Collapse
|
8
|
Zhu P, Tao H, Chen K, Chu M, Wang Q, Yang X, Zhou J, Yang H, Geng D. TRPA1 aggravates osteoclastogenesis and osteoporosis through activating endoplasmic reticulum stress mediated by SRXN1. Cell Death Dis 2024; 15:624. [PMID: 39191723 DOI: 10.1038/s41419-024-07018-5] [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: 05/23/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Osteoporosis (OP) is a disorder of bone remodeling caused by an imbalance between bone resorption by osteoclasts and bone formation by osteoblasts. Therefore, inhibiting excessive osteoclast activity is one of the promising strategies for treating OP. A major transient receptor potential cation channel, known as transient receptor potential ankyrin 1 (TRPA1), was found to alleviate joint pain and cartilage degeneration in osteoarthritis. However, little research has focused on TRPA1 function in OP. As a result, this study aimed to explore the TRPA1 characteristics and its potential therapeutic function during osteoclastogenesis. The TRPA1 expression gradually increased in the osteoclast differentiation process; however, its suppression with small interfering RNA and an inhibitor (HC030031) significantly controlled the osteoclast count and the expression of osteoclast characteristic genes. Its suppression also inhibited endoplasmic reticulum (ER) stress-related pancreatic ER kinase (PERK) pathways. An ER stress inhibitor (thapsigargin) reversed the down-regulated levels of ER stress and osteoclast differentiation by suppressing TRPA1. Transcriptome sequencing results demonstrated that TRPA1 negatively regulated reactive oxygen species (ROS) and significantly increased the expression of an antioxidant gene, SRXN1. The osteoclast differentiation and the levels of ER stress were enhanced with SRXN1 inhibition. Finally, TRPA1 knockdown targeting macrophages by adeno-associated virus-9 could relieve osteoclast differentiation and osteopenia in ovariectomized mice. In summary, silencing TRPA1 restrained osteoclast differentiation through ROS-mediated down-regulation of ER stress via inhibiting PERK pathways. The study also indicated that TRPA1 might become a prospective treatment target for OP.
Collapse
Affiliation(s)
- Pengfei Zhu
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Kai Chen
- Department of Orthopedics, Hai'an People's Hospital, Hai'an, 226600, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, Yixing People's Hospital, Yixing, 214200, Jiangsu, China
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, 215500, Jiangsu, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215008, Jiangsu, China.
| | - Jun Zhou
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
9
|
Herbert A. Osteogenesis imperfecta type 10 and the cellular scaffolds underlying common immunological diseases. Genes Immun 2024; 25:265-276. [PMID: 38811682 DOI: 10.1038/s41435-024-00277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
Osteogenesis imperfecta type 10 (OI10) is caused by loss of function codon variants in the gene SERPINH1 that encodes heat shock protein 47 (HSP47), rather than in a gene specifying bone formation. The HSP47 variants disrupt the folding of both collagen and the endonuclease IRE1α (inositol-requiring enzyme 1α) that splices X-Box Binding Protein 1 (XBP1) mRNA. Besides impairing bone development, variants likely affect osteoclast differentiation. Three distinct biochemical scaffold play key roles in the differentiation and regulated cell death of osteoclasts. These scaffolds consist of non-templated protein modifications, ordered lipid arrays, and protein filaments. The scaffold components are specified genetically, but assemble in response to extracellular perturbagens, pathogens, and left-handed Z-RNA helices encoded genomically by flipons. The outcomes depend on interactions between RIPK1, RIPK3, TRIF, and ZBP1 through short interaction motifs called RHIMs. The causal HSP47 nonsynonymous substitutions occur in a novel variant leucine repeat region (vLRR) that are distantly related to RHIMs. Other vLRR protein variants are causal for a variety of different mendelian diseases. The same scaffolds that drive mendelian pathology are associated with many other complex disease outcomes. Their assembly is triggered dynamically by flipons and other context-specific switches rather than by causal, mendelian, codon variants.
Collapse
Affiliation(s)
- Alan Herbert
- InsideOutBio, 42 8th Street, Charlestown, MA, USA.
| |
Collapse
|
10
|
Materozzi M, Resnati M, Facchi C, Trudu M, Orfanelli U, Perini T, Gennari L, Milan E, Cenci S. A novel proteomic signature of osteoclast differentiation unveils the deubiquitinase UCHL1 as a necessary osteoclastogenic driver. Sci Rep 2024; 14:7290. [PMID: 38538704 PMCID: PMC10973525 DOI: 10.1038/s41598-024-57898-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024] Open
Abstract
Bone destruction, a major source of morbidity, is mediated by heightened differentiation and activity of osteoclasts (OC), highly specialized multinucleated myeloid cells endowed with unique bone-resorptive capacity. The molecular mechanisms regulating OC differentiation in the bone marrow are still partly elusive. Here, we aimed to identify new regulatory circuits and actionable targets by comprehensive proteomic characterization of OCgenesis from mouse bone marrow monocytes, adopting two parallel unbiased comparative proteomic approaches. This work disclosed an unanticipated protein signature of OCgenesis, with most gene products currently unannotated in bone-related functions, revealing broad structural and functional cellular reorganization and divergence from macrophagic immune activity. Moreover, we identified the deubiquitinase UCHL1 as the most upregulated cytosolic protein in differentiating OCs. Functional studies proved it essential, as UCHL1 genetic and pharmacologic inhibition potently suppressed OCgenesis. Furthermore, proteomics and mechanistic dissection showed that UCHL1 supports OC differentiation by restricting the anti-OCgenic activity of NRF2, the transcriptional activator of the canonical antioxidant response, through redox-independent stabilization of the NRF2 inhibitor, KEAP1. Besides offering a valuable experimental framework to dissect OC differentiation, our study discloses the essential role of UCHL1, exerted through KEAP1-dependent containment of NRF2 anti-OCgenic activity, yielding a novel potential actionable pathway against bone loss.
Collapse
Affiliation(s)
- Maria Materozzi
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Massimo Resnati
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cecilia Facchi
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Matteo Trudu
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Ugo Orfanelli
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Perini
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Enrico Milan
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Simone Cenci
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
11
|
Zhao S, Zhao H, Zhao L, Cheng X, Zheng Z, Wu M, Wen W, Wang S, Zhou Z, Xie H, Ruan D, Li Q, Liu X, Ou C, Li G, Zhao Z, Chen G, Niu Y, Yin X, Hu Y, Zhang X, Liu P, Qiu G, Liu W, Zhao C, Wu Z, Zhang J, Wu N. Unraveling the genetic architecture of congenital vertebral malformation with reference to the developing spine. Nat Commun 2024; 15:1125. [PMID: 38321032 PMCID: PMC10847475 DOI: 10.1038/s41467-024-45442-5] [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: 09/02/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024] Open
Abstract
Congenital vertebral malformation, affecting 0.13-0.50 per 1000 live births, has an immense locus heterogeneity and complex genetic architecture. In this study, we analyze exome/genome sequencing data from 873 probands with congenital vertebral malformation and 3794 control individuals. Clinical interpretation identifies Mendelian etiologies in 12.0% of the probands and reveals a muscle-related disease mechanism. Gene-based burden test of ultra-rare variants identifies risk genes with large effect sizes (ITPR2, TBX6, TPO, H6PD, and SEC24B). To further investigate the biological relevance of the genetic association signals, we perform single-nucleus RNAseq on human embryonic spines. The burden test signals are enriched in the notochord at early developmental stages and myoblast/myocytes at late stages, highlighting their critical roles in the developing spine. Our work provides insights into the developmental biology of the human spine and the pathogenesis of spine malformation.
Collapse
Affiliation(s)
- Sen Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hengqiang Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lina Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xi Cheng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhifa Zheng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Mengfan Wu
- Institute of Evolution & Marine Biodiversity, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Wen Wen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shengru Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zixiang Zhou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Haibo Xie
- Institute of Evolution & Marine Biodiversity, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Dengfeng Ruan
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Road, Haining, 314400, China
| | - Qing Li
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinquan Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Chengzhu Ou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Guozhuang Li
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhengye Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Guilin Chen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuchen Niu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangjie Yin
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuhong Hu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaochen Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Baylor Genetics, Houston, TX, 77021, USA
| | - Guixing Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Wanlu Liu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Road, Haining, 314400, China
| | - Chengtian Zhao
- Institute of Evolution & Marine Biodiversity, College of Marine Life Science, Ocean University of China, Qingdao, 266003, China
| | - Zhihong Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Jianguo Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Nan Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, 100730, China.
- Key laboratory of big data for spinal deformities, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
12
|
Ahmad P, Siqueira WL. Polymorphism of salivary proteins and risk of periodontal diseases: A systematic review and meta-analysis of clinical studies. J Dent 2024; 141:104804. [PMID: 38122885 DOI: 10.1016/j.jdent.2023.104804] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/17/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023] Open
Abstract
OBJECTIVES The present systematic review and meta-analysis aimed to assess the association between salivary protein polymorphisms and the risk of periodontal diseases (PD). DATA The review incorporated cross-sectional, case-control, retrospective/prospective cohort, and randomized controlled trials assessing the influence of salivary protein polymorphisms on the risk of PD development were included in this review. SOURCES A thorough literature search was conducted across electronic databases, namely PubMed, Scopus, Embase, and Web of Science, without any restrictions on publication language and year. STUDY SELECTION A total of 168 studies were identified, of which 19 were eligible for inclusion. The risk of bias (RoB) assessment of the included studies was conducted at the methodological level. RESULTS A total of 16 studies were included. Polymorphism in the gene encoding TNF-α was found to be protective against gingivitis, while those encoding IL-1α and IL-1β were associated with developing gingivitis. Of the 42 proteins investigated, various gene polymorphisms were identified as protective or risk factors for periodontitis. Protective genes include CFH, DNMT1, OPRM1, and TLR9. Conversely, certain salivary protein genes (e.g., CRP, ERN1, FAM5C, IDH2, LTA, TET2, MPA, NLRP3, TLR4) were associated with periodontitis risk. Notably, IL6, MMP9, and MUC7 genes showed no association with PD, while MMP13 was linked to early implant loss. Overall, the meta-analysis found a statistically significant association between salivary proteins' polymorphisms and risk of PD. CONCLUSIONS Salivary protein polymorphisms significantly influence PD, revealing protective and risk-associated genotypes. Despite limitations, findings suggest therapeutic targets, emphasizing the complex genetics-periodontal health interplay. CLINICAL SIGNIFICANCE This study unveils salivary protein polymorphisms as pivotal factors in PD. Protective genes including CFH and TLR9, and risk-associated genes including CRP and TLR4, indicate a genetic basis for PD susceptibility.
Collapse
Affiliation(s)
- Paras Ahmad
- College of Dentistry, University of Saskatchewan, Saskatoon, S7N E5E, Saskatchewan, Canada
| | - Walter Luiz Siqueira
- College of Dentistry, University of Saskatchewan, Saskatoon, S7N E5E, Saskatchewan, Canada.
| |
Collapse
|
13
|
Liang L, Zhang F, Feng N, Kuang B, Fan M, Chen C, Pan Y, Zhou P, Geng N, Li X, Xian M, Deng L, Li X, Kuang L, Luo F, Tan Q, Xie Y, Guo F. IRE1α protects against osteoarthritis by regulating progranulin-dependent XBP1 splicing and collagen homeostasis. Exp Mol Med 2023; 55:2376-2389. [PMID: 37907740 PMCID: PMC10689778 DOI: 10.1038/s12276-023-01106-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/17/2023] [Accepted: 08/02/2023] [Indexed: 11/02/2023] Open
Abstract
Osteoarthritis (OA) is a full-joint, multifactorial, degenerative and inflammatory disease that seriously affects the quality of life of patients due to its disabling and pain-causing properties. ER stress has been reported to be closely related to the progression of OA. The inositol-requiring enzyme 1α/X-box-binding protein-1 spliced (IRE1α/XBP1s) pathway, which is highly expressed in the chondrocytes of OA patients, promotes the degradation and refolding of abnormal proteins during ER stress and maintains the stability of the ER environment of chondrocytes, but its function and the underlying mechanisms of how it contributes to the progression of OA remain unclear. This study investigates the role of IRE1α/ERN1 in OA. Specific deficiency of ERN1 in chondrocytes spontaneously resulted in OA-like cartilage destruction and accelerated OA progression in a surgically induced arthritis model. Local delivery of AdERN1 relieved degradation of the cartilage matrix and prevented OA development in an ACLT-mediated model. Mechanistically, progranulin (PGRN), an intracellular chaperone, binds to IRE1α, promoting its phosphorylation and splicing of XBP1u to generate XBP1s. XBP1s protects articular cartilage through TNF-α/ERK1/2 signaling and further maintains collagen homeostasis by regulating type II collagen expression. The chondroprotective effect of IRE1α/ERN1 is dependent on PGRN and XBP1s splicing. ERN1 deficiency accelerated cartilage degeneration in OA by reducing PGRN expression and XBP1s splicing, subsequently decreasing collagen II expression and triggering collagen structural abnormalities and an imbalance in collagen homeostasis. This study provides new insights into OA pathogenesis and the UPR and suggests that IRE1α/ERN1 may serve as a potential target for the treatment of joint degenerative diseases, including OA.
Collapse
Affiliation(s)
- Li Liang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Fengmei Zhang
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
- Laboratory Animal Center, Chongqing Medical University, 400016, Chongqing, China
| | - Naibo Feng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Biao Kuang
- Department of Orthopedics, The 2nd Affiliated Hospital of Chongqing Medical University, 400072, Chongqing, China
| | - Mengtian Fan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Cheng Chen
- Department of Orthopedics, The 1st Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Yiming Pan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Pengfei Zhou
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, College of Stomatology, Chongqing Medical University, 400016, Chongqing, China
| | - Nana Geng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Xingyue Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Menglin Xian
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Lin Deng
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoli Li
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
| | - Liang Kuang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Fengjin Guo
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
14
|
Lv W, Zheng Y, Jiao J, Fu Y, Xu T, Zhang L, Zhang Z, Ma N. The Role of XBP1 in bone metabolism. Front Endocrinol (Lausanne) 2023; 14:1217579. [PMID: 37795354 PMCID: PMC10546391 DOI: 10.3389/fendo.2023.1217579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
Bone is a dynamic organ that, once formed, undergoes a constant remodeling process that includes bone resorption and synthesis. Osteoclasts and osteoblasts are primarily responsible for controlling this process. X-box binding protein 1 (XBP1), a transcription factor, affects the metabolism of bones in various ways. In recent years, numerous studies have revealed that XBP1 plays a vital role in bone metabolism, including osteoclast and osteoblast development, as well as in regulating immune cell differentiation that affects the immune microenvironment of bone remodeling. In this review, we highlight the regulatory mechanisms of XBP1 on osteoclasts and osteoblasts, how XBP1 affects the immune microenvironment of bone remodeling by influencing the differentiation of immune cells, and predict the possible future research directions of XBP1 to provide new insights for the treatment of bone-related metabolic diseases.
Collapse
Affiliation(s)
- Wenhao Lv
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Youli Zheng
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Junjun Jiao
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Fu
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Tingrui Xu
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Li Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Zheng Zhang
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China
| | - Ning Ma
- Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
15
|
Zhong M, Wu Z, Chen Z, Ren Q, Zhou J. Advances in the interaction between endoplasmic reticulum stress and osteoporosis. Biomed Pharmacother 2023; 165:115134. [PMID: 37437374 DOI: 10.1016/j.biopha.2023.115134] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
The endoplasmic reticulum (ER) is the main site for protein synthesis, folding, and secretion, and accumulation of the unfolded/misfolded proteins in the ER may induce ER stress. ER stress is an important participant in various intracellular signaling pathways. Prolonged- or high-intensity ER stress may induce cell apoptosis. Osteoporosis, characterized by imbalanced bone remodeling, is a global disease caused by many factors, such as ER stress. ER stress stimulates osteoblast apoptosis, increases bone loss, and promotes osteoporosis development. Many factors, such as the drug's adverse effects, metabolic disorders, calcium ion imbalance, bad habits, and aging, have been reported to activate ER stress, resulting in the pathological development of osteoporosis. Increasing evidence shows that ER stress regulates osteogenic differentiation, osteoblast activity, and osteoclast formation and function. Various therapeutic agents have been developed to counteract ER stress and thereby suppress osteoporosis development. Thus, inhibition of ER stress has become a potential target for the therapeutic management of osteoporosis. However, the in-depth understanding of ER stress in the pathogenesis of osteoporosis still needs more effort.
Collapse
Affiliation(s)
- Mingliang Zhong
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China
| | - Zhenyu Wu
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Qun Ren
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Blocking the cytohesin-2/ARF1 axis by SecinH3 ameliorates osteoclast-induced bone loss via attenuating JNK-mediated IRE1 endoribonuclease activity. Pharmacol Res 2022; 185:106513. [DOI: 10.1016/j.phrs.2022.106513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/19/2022]
|
19
|
Wang G, Fan F, Sun C, Hu Y. Looking into Endoplasmic Reticulum Stress: The Key to Drug-Resistance of Multiple Myeloma? Cancers (Basel) 2022; 14:5340. [PMID: 36358759 PMCID: PMC9654020 DOI: 10.3390/cancers14215340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 09/22/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, resulting from the clonal proliferation of malignant plasma cells within the bone marrow. Despite significant advances that have been made with novel drugs over the past two decades, MM patients often develop therapy resistance, especially to bortezomib, the first-in-class proteasome inhibitor that was approved for treatment of MM. As highly secretory monoclonal protein-producing cells, MM cells are characterized by uploaded endoplasmic reticulum stress (ERS), and rely heavily on the ERS response for survival. Great efforts have been made to illustrate how MM cells adapt to therapeutic stresses through modulating the ERS response. In this review, we summarize current knowledge on the mechanisms by which ERS response pathways influence MM cell fate and response to treatment. Moreover, based on promising results obtained in preclinical studies, we discuss the prospect of applying ERS modulators to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Guangqi Wang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
| | - Fengjuan Fan
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
| | - Chunyan Sun
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
20
|
Liang W, Chen Q, Cheng S, Wei R, Li Y, Yao C, Ouyang Z, Kang D, Chen A, Liu Z, Li K, Bai X, Li Q, Huang B. Skin chronological aging drives age-related bone loss via secretion of cystatin-A. NATURE AGING 2022; 2:906-922. [PMID: 37118283 DOI: 10.1038/s43587-022-00285-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 08/25/2022] [Indexed: 04/30/2023]
Abstract
Although clinical evidence has indicated an association between skin atrophy and bone loss during aging, their causal relationship and the underlying mechanisms are unknown. Here we show that premature skin aging drives bone loss in mice. We further identify that cystatin-A (Csta), a keratinocyte-enriched secreted factor, mediates the effect of skin on bone. Keratinocyte-derived Csta binds the receptor for activated C-kinase 1 in osteoblast and osteoclast progenitors, thus promoting their proliferation but inhibiting osteoclast differentiation. Csta secretion decreases with skin aging in both mice and humans, thereby causing senile osteoporosis by differentially decreasing the numbers of osteoblasts and osteoclasts. In contrast, topical application of calcipotriol stimulates Csta production in the epidermis and alleviates osteoporosis. These results reveal a mode of endocrine regulation of bone metabolism in the skin, and identify Csta as an epidermally derived hormone linking skin aging to age-related bone loss. Enhancers of skin Csta levels could serve as a potential topical drug for treatment of senile osteoporosis.
Collapse
Affiliation(s)
- Wenquan Liang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qingjing Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shasha Cheng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ruiming Wei
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yuejun Li
- Department of Orthopedics, The Second People's Hospital of Panyu District, Guangzhou, China
| | - Chenfeng Yao
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhicong Ouyang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dawei Kang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ajuan Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zezheng Liu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Kai Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Qingchu Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Bin Huang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
21
|
Jing G, Zuo J, Fang Q, Yuan M, Xia Y, Jin Q, Liu Y, Wang Y, Zhang Z, Liu W, Wu X, Song X. Erbin protects against sepsis-associated encephalopathy by attenuating microglia pyroptosis via IRE1α/Xbp1s-Ca 2+ axis. J Neuroinflammation 2022; 19:237. [PMID: 36171629 PMCID: PMC9520943 DOI: 10.1186/s12974-022-02598-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/16/2022] [Indexed: 06/27/2024] Open
Abstract
Background Microglia pyroptosis-mediated neuroinflammation is thought to be the crucial pathogenesis of sepsis-associated encephalopathy (SAE). Erbin has been reported to be associated with various inflammatory diseases. However, the role of Erbin in SAE and the relationship between Erbin and microglia pyroptosis are unknown. In this study, we investigated the promising role and underlying molecular mechanism of Erbin in the regulation of microglia pyroptosis. Methods WT and Erbin knockout mice underwent cecum ligation perforation (CLP) to induce SAE. Primary mouse microglia and BV2 cells were treated with LPS/nigericin in vitro. Behavioral tests were performed to evaluate cognitive function. Nissl staining and transmission electron microscopy were used to assess histological and structural lesions. ELISA and qPCR were carried out to detect neuroinflammation. Western blot and immunofluorescence were used to analyze protein expression. Flow cytometry and confocal microscopy were utilized to observe the Ca2+ changes in the cytoplasm and endoplasmic reticulum (ER). To further explore the underlying mechanism, STF083010 was administered to block the IRE1α/Xbp1s pathway. Results Erbin deletion resulted in more pronounced neuronal damage and cognitive impairment in mice that underwent CLP. Erbin knockout promoted microglial pyroptosis and inflammatory cytokines secretion in vivo and in vitro, which was mediated by activation of the IRE1α/Xbp1s. Treatment with the selective inhibitor STF083010 significantly inhibited IRE1α/Xbp1s pathway activity, decreased intracytoplasmic Ca2+, attenuated microglial pyroptosis, reduced pro-inflammatory cytokine secretion, lessened neuronal damage, and improved cognitive function. Conclusions In SAE, Erbin inhibits IRE1/Xbp1s pathway activity and reduces the ER Ca2+ influx to the cytoplasm, reducing microglial pyroptosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02598-5.
Collapse
Affiliation(s)
- Guoqing Jing
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Zuo
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qing Fang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yun Xia
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiyan Jin
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuping Liu
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanlin Wang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zongze Zhang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wanhong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
22
|
Turishcheva E, Vildanova M, Onishchenko G, Smirnova E. The Role of Endoplasmic Reticulum Stress in Differentiation of Cells of Mesenchymal Origin. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:916-931. [PMID: 36180988 PMCID: PMC9483250 DOI: 10.1134/s000629792209005x] [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: 05/05/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 05/23/2023]
Abstract
Endoplasmic reticulum (ER) is a multifunctional membrane-enclosed organelle. One of the major ER functions is cotranslational transport and processing of secretory, lysosomal, and transmembrane proteins. Impaired protein processing caused by disturbances in the ER homeostasis results in the ER stress. Restoration of normal ER functioning requires activation of an adaptive mechanism involving cell response to misfolded proteins, the so-called unfolded protein response (UPR). Besides controlling protein folding, UPR plays a key role in other physiological processes, in particular, differentiation of cells of connective, muscle, epithelial, and neural tissues. Cell differentiation is induced by the physiological levels of ER stress, while excessive ER stress suppresses differentiation and can result in cell death. So far, it remains unknown whether UPR activation induces cell differentiation or if UPR is initiated by the upregulated synthesis of secretory proteins during cell differentiation. Cell differentiation is an important stage in the development of multicellular organisms and is tightly controlled. Suppression or excessive activation of this process can lead to the development of various pathologies in an organism. In particular, impairments in the differentiation of connective tissue cells can result in the development of fibrosis, obesity, and osteoporosis. Recently, special attention has been paid to fibrosis as one of the major complications of COVID-19. Therefore, studying the role of UPR in the activation of cell differentiation is of both theoretical and practical interest, as it might result in the identification of molecular targets for selective regulation of cell differentiation stages and as well as the potential to modulate the mechanisms involved in the development of various pathological states.
Collapse
Affiliation(s)
| | - Mariya Vildanova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Galina Onishchenko
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elena Smirnova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
23
|
Duxfield A, Munkley J, Briggs MD, Dennis EP. CRELD2 is a novel modulator of calcium release and calcineurin-NFAT signalling during osteoclast differentiation. Sci Rep 2022; 12:13884. [PMID: 35974042 PMCID: PMC9381524 DOI: 10.1038/s41598-022-17347-0] [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: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022] Open
Abstract
Cysteine rich with epidermal growth factor (EGF)-like domains 2 (CRELD2) is an endoplasmic reticulum (ER) resident chaperone protein with calcium binding properties. CRELD2 is an ER-stress regulated gene that has been implicated in the pathogenesis of skeletal dysplasias and has been shown to play an important role in the differentiation of chondrocytes and osteoblasts. Despite CRELD2 having an established role in skeletal development and bone formation, its role in osteoclasts is currently unknown. Here we show for the first time that CRELD2 plays a novel role in trafficking transforming growth factor beta 1 (TGF-β1), which is linked to an upregulation in the expression of Nfat2, the master regulator of osteoclast differentiation in early osteoclastogenesis. Despite this finding, we show that overexpressing CRELD2 impaired osteoclast differentiation due to a reduction in the activity of the calcium-dependant phosphatase, calcineurin. This in turn led to a subsequent block in the dephosphorylation of nuclear factor of activated T cells 1 (NFATc1), preventing its nuclear localisation and activation as a pro-osteoclastogenic transcription factor. Our exciting results show that the overexpression of Creld2 in osteoclasts impaired calcium release from the ER which is essential for activating calcineurin and promoting osteoclastogenesis. Therefore, our data proposes a novel inhibitory role for this calcium-binding ER-resident chaperone in modulating calcium flux during osteoclast differentiation which has important implications in our understanding of bone remodelling and the pathogenesis of skeletal diseases.
Collapse
Affiliation(s)
- Adam Duxfield
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Jennifer Munkley
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Michael D Briggs
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK
| | - Ella P Dennis
- International Centre for Life, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
24
|
Yang P, Zhang T, Zhu R, Shen Y, Pan Y, Zhang Y. Protective effect of zinc supplementation on tricalcium phosphate particles-induced inflammatory osteolysis in mice. Microsc Res Tech 2022; 85:3608-3617. [PMID: 35876446 DOI: 10.1002/jemt.24213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022]
Abstract
Zinc (Zn), an essential trace element, can stimulate bone formation and inhibit osteoclastic bone resorption, which controls the growth and maintenance of bone. However, the effect of Zn supplementation on tricalcium phosphate (TCP) wear particles-induced osteolysis remains unknown. Here, we doped Zn into TCP particles (ZnTCP), and explore the protective effects of Zn on TCP particles-induced osteolysis in vivo. TCP particles and ZnTCP particles were embedded under the periosteum around the middle suture of the mouse calvaria. After 2 weeks, blood, the periosteal tissue, and the calvaria were collected to determine serum levels of Zn and osteocalcin, pro-inflammatory cytokines, bone biochemical markers, osteoclastogenesis and bone resorption area, and to explain its mechanism. Data revealed that Zn significantly prevented TCP particles-induced osteoclastogenesis and bone loss, and increased bone turnover. The Zn supplement remarkably suppressed the release of pro-inflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6. Immunoblotting demonstrated that Zn alleviated expression levels of ER stress-related proteins such as glucose-regulated protein 78 (GRP78), PKR-like ER kinase (PERK), phospho-PERK (p-PERK), eukaryotic initiation factor 2α (eIF2α), phospho-eIF2α (p-eIF2α), activating transcription factor 4 (ATF4), inositol-requiring enzyme 1α (IRE1-α) and transcription factor X-box binding protein spliced (XBP1s), leading to decreasing the ratios of p-PERK/PERK and p-eIF2α/eIF2α. Taken together, Zn supplementation strongly prevents TCP particles-induced periprosthetic osteolysis via inhibition of the ER stress pathway, and it may be a novel therapeutic approach for the treatment of aseptic prosthesis loosening.
Collapse
Affiliation(s)
- Pei Yang
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Tao Zhang
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Ruirong Zhu
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Yuchen Shen
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Yuefang Pan
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Yun Zhang
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| |
Collapse
|
25
|
Chiu HW, Hou YC, Lu CL, Lu KC, Liu WC, Shyu JF, Chang JF, Zheng CM. Cinacalcet Improves Bone Parameters Through Regulation of Osteoclast Endoplasmic Reticulum Stress, Autophagy, and Apoptotic Pathways in Chronic Kidney Disease-Mineral and Bone Disorder. J Bone Miner Res 2022; 37:215-225. [PMID: 34633122 DOI: 10.1002/jbmr.4459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 09/13/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022]
Abstract
The possible mechanisms underlying the quantitative and qualitative effects of cinacalcet on bone were explored in a chronic kidney disease-mineral and bone disorder (CKD-MBD) mouse model in relation to the influence of the interactions among the osteoclast (OC) endoplasmic reticulum (ER) stress, autophagy and apoptosis pathways on OC differentiation. Body weight and biochemical parameters improved significantly in the CKD + cinacalcet groups compared to the CKD group. Micro-computed tomography (μCT) revealed both cortical and trabecular parameters deteriorated significantly in the CKD group and were reversed by cinacalcet in a dose-dependent manner. Nanoindentation analysis of bone quality proved that both cortical hardness and elastic modulus improved significantly with high dose cinacalcet treatment. In vitro studies revealed that cinacalcet inhibited receptor activator of NF-κB ligand (RANKL)/receptor activator of NF-κB (RANK)-induced OC differentiation in a concentration-dependent manner through a close interaction between activation of caspase-related apoptosis, reversal of OC autophagy through the protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK) pathways, and attenuation of the OC ER stress/CREBH/NFATc1 signaling pathway. Cinacalcet improves both bone quantity and bone quality in CKD mouse model and inhibits OC differentiation through regulation of the interactions among the apoptosis, ER stress, and autophagy pathways within OCs. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,TMU Research Centre of Urology and Kidney, Taipei Medical University, Taipei, Taiwan.,Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Chou Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, New Taipei City, Taiwan.,School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chien-Lin Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.,Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Kuo-Cheng Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan.,Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan.,Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan
| | - Wen-Chih Liu
- Division of Nephrology, Department of Internal Medicine, Taipei Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan.,Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jia-Fwu Shyu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Jia-Feng Chang
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,TMU Research Centre of Urology and Kidney, Taipei Medical University, Taipei, Taiwan.,Department of Nursing, Yuanpei University of Medical Technology, Hsinchu, Taiwan.,Division of Nephrology, Department of Internal Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,TMU Research Centre of Urology and Kidney, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
26
|
Mi B, Chen L, Xiong Y, Yang Y, Panayi AC, Xue H, Hu Y, Yan C, Hu L, Xie X, Lin Z, Zhou W, Cao F, Xiao X, Feng Q, Liu G. Osteoblast/Osteoclast and Immune Cocktail Therapy of an Exosome/Drug Delivery Multifunctional Hydrogel Accelerates Fracture Repair. ACS NANO 2022; 16:771-782. [PMID: 34979087 DOI: 10.1021/acsnano.1c08284] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The osteoblast/osteoclast and M1/M2 macrophage ratios play critical roles in delayed fracture healing. Robust osteoblast differentiation and M2 macrophage polarization can substantiality promote fracture repair; however, the combined effect of these strategies has not been previously studied. In this study, we constructed a cocktail therapy to simultaneously regulate the osteoblast/osteoclast and M1/M2 macrophage balance. The cocktail therapy composed of a natural polymer hyaluronic-acid-based hydrogel (HA hydrogel, which has a tissue-adhesive, injectable, self-healing, anti-inflammation profile), engineered endothelial cell-derived exosomes (EC-ExosmiR-26a-5p), and APY29, an IRE-1α inhibitor. This allowed for specific delivery of EC-ExosmiR-26a-5p and APY29 for osteoblast/osteoclast and macrophage regulation, respectively. The results suggested that the cocktail therapy exerted pro-fracture repair effects with each of its components established as indispensable. The assessed cocktail therapy provides insight into synergistic strategies and is useful for developing more suitable pro-fracture repair therapy.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yayan Yang
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou 350007, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School Boston, Massachusetts 02152, United States
| | - Hang Xue
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yiqiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Chenchen Yan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Liangcong Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xudong Xie
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiufeng Xiao
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou 350007, China
| | - Qian Feng
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou 350007, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
27
|
Li Q, Duan Z, Sun C, Zheng J, Xu G, Yang N. Genetic variations for the eggshell crystal structure revealed by genome-wide association study in chickens. BMC Genomics 2021; 22:786. [PMID: 34727889 PMCID: PMC8565016 DOI: 10.1186/s12864-021-08103-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/18/2021] [Indexed: 11/10/2022] Open
Abstract
Background Eggshell is a bio-ceramic material comprising columnar calcite (CaCO3) crystals and organic proteinaceous matrix. The size, shape and orientation of the CaCO3 crystals influence the microstructural properties of chicken eggshells. However, the genetic architecture underlying eggshell crystal polymorphism remains to be elucidated. Results The integral intensity of the nine major diffraction peaks, total integral intensity and degree of orientation of the crystals were measured followed by a genome-wide association study in 839 F2 hens. The results showed that the total integral intensity was positively correlated with the eggshell strength, eggshell thickness, eggshell weight, mammillary layer thickness and effective layer thickness. The SNP-based heritabilities of total integral intensity and degree of orientation were 0.23 and 0.06, respectively. The 621 SNPs located in the range from 55.6 to 69.1 Mb in GGA1 were significantly associated with TA. PLCZ1, ABCC9, ITPR2, KCNJ8, CACNA1C and IAPP, which are involved in the biological process of regulating cytosolic calcium ion concentration, can be suggested as key genes regulating the total integral intensity. Conclusions The findings greatly advance the understanding of the genetic basis underlying the crystal ultrastructure of eggshell quality and thus will have practical significance in breeding programs for improving eggshell quality. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08103-1.
Collapse
Affiliation(s)
- Quanlin Li
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Zhongyi Duan
- National Animal Husbandry Service, 100125, Beijing, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Jiangxia Zheng
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Guiyun Xu
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
28
|
Herlea-Pana O, Eeda V, Undi RB, Lim HY, Wang W. Pharmacological Inhibition of Inositol-Requiring Enzyme 1α RNase Activity Protects Pancreatic Beta Cell and Improves Diabetic Condition in Insulin Mutation-Induced Diabetes. Front Endocrinol (Lausanne) 2021; 12:749879. [PMID: 34675883 PMCID: PMC8524045 DOI: 10.3389/fendo.2021.749879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
β-cell ER stress plays an important role in β-cell dysfunction and death during the pathogenesis of diabetes. Proinsulin misfolding is regarded as one of the primary initiating factors of ER stress and unfolded protein response (UPR) activation in β-cells. Here, we found that the ER stress sensor inositol-requiring enzyme 1α (IRE1α) was activated in the Akita mice, a mouse model of mutant insulin gene-induced diabetes of youth (MIDY), a monogenic diabetes. Normalization of IRE1α RNase hyperactivity by pharmacological inhibitors significantly ameliorated the hyperglycemic conditions and increased serum insulin levels in Akita mice. These benefits were accompanied by a concomitant protection of functional β-cell mass, as shown by the suppression of β-cell apoptosis, increase in mature insulin production and reduction of proinsulin level. At the molecular level, we observed that the expression of genes associated with β-cell identity and function was significantly up-regulated and ER stress and its associated inflammation and oxidative stress were suppressed in islets from Akita mice treated with IRE1α RNase inhibitors. This study provides the evidence of the in vivo efficacy of IRE1α RNase inhibitors in Akita mice, pointing to the possibility of targeting IRE1α RNase as a therapeutic direction for the treatment of diabetes.
Collapse
Affiliation(s)
- Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Hui-Ying Lim
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, Oklahoma City, OK, United States
| |
Collapse
|
29
|
Zhang J, Zhang J, Ni H, Wang Y, Katwal G, Zhao Y, Sun K, Wang M, Li Q, Chen G, Miao Y, Gong N. Downregulation of XBP1 protects kidney against ischemia-reperfusion injury via suppressing HRD1-mediated NRF2 ubiquitylation. Cell Death Discov 2021; 7:44. [PMID: 33654072 PMCID: PMC7925512 DOI: 10.1038/s41420-021-00425-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
Ischemia-reperfusion (IR) injury to the renal epithelia is associated with endoplasmic reticulum stress (ERS) and mitochondria dysfunction, which lead to oxidative stress-induced acute kidney injury (AKI). X-box binding protein 1 (XBP1), an ERS response protein, could play a prominent role in IR-induced AKI. In this study, we revealed that XBP1 and its downstream target HRD1 participated in the crosstalk between ERS and mitochondrial dysfunction via regulation of NRF2/HO-1-mediated reactive oxidative stress (ROS) signaling. Mice with reduced expression of XBP1 (heterozygous Xbp1±) were resistant to IR-induced AKI due to the enhanced expression of NRF2/HO-1 and diminished ROS in the kidney. Downregulation of XBP1 in renal epithelial cells resulted in reduced HRD1 expression and increased NRF2/HO-1 function, accompanied with enhanced antioxidant response. Furthermore, HRD1 served as an E3-ligase to facilitate the downregulation of NRF2 through ubiquitination-degradation pathway, and the QSLVPDI motif on NRF2 constituted an active site for its interaction with HRD1. Thus, our findings unveil an important physiological role for XBP1/HRD1 in modulating the antioxidant function of NRF2/HO-1 in the kidney under stress conditions. Molecular therapeutic approaches that target XBP1-HRD1-NRF2 pathway may represent potential effective means to treat renal IR injury.
Collapse
Affiliation(s)
- Ji Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China
| | - Jiasi Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China
| | - Haiqiang Ni
- Organ Transplant Department, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Yanfeng Wang
- Institute of Hepatobiliary Diseases, Transplant Center, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital, Wuhan University, 430071, Wuhan, Hubei, China
| | - Gaurav Katwal
- Chitwan Medical College Teaching Hospital, Department of Surgery, Bharatpur, Chitwan, 44200, Nepal
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China
| | - Kailun Sun
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China
| | - Mengqin Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China
| | - Qingwen Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China
| | - Gen Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, Hubei, China
| | - Yun Miao
- Organ Transplant Department, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Nianqiao Gong
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, 430030, Wuhan, Hubei, China.
| |
Collapse
|
30
|
Zhang D, De Veirman K, Fan R, Jian Q, Zhang Y, Lei L, Evans H, Wang Y, Lei L, Wang B, Williamson RA, Chantry A, He P, Li A, De Raeve H, Vanderkerken K, He A, Hu J. ER stress arm XBP1s plays a pivotal role in proteasome inhibition-induced bone formation. Stem Cell Res Ther 2020; 11:516. [PMID: 33256835 PMCID: PMC7708206 DOI: 10.1186/s13287-020-02037-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Bone destruction is a hallmark of multiple myeloma (MM). It has been reported that proteasome inhibitors (PIs) can reduce bone resorption and increase bone formation in MM patients, but the underlying mechanisms remain unclear. METHODS Mesenchymal stem cells (MSCs) were treated with various doses of PIs, and the effects of bortezomib or carfilzomib on endoplasmic reticulum (ER) stress signaling pathways were analyzed by western blotting and real-time PCR. Alizarin red S (ARS) and alkaline phosphatase (ALP) staining were used to determine the osteogenic differentiation in vitro. Specific inhibitors targeting different ER stress signaling and a Tet-on inducible overexpressing system were used to validate the roles of key ER stress components in regulating osteogenic differentiation of MSCs. Chromatin immunoprecipitation (ChIP) assay was used to evaluate transcription factor-promoter interaction. MicroCT was applied to measure the microarchitecture of bone in model mice in vivo. RESULTS We found that both PERK-ATF4 and IRE1α-XBP1s ER stress branches are activated during PI-induced osteogenic differentiation. Inhibition of ATF4 or XBP1s signaling can significantly impair PI-induced osteogenic differentiation. Furthermore, we demonstrated that XBP1s can transcriptionally upregulate ATF4 expression and overexpressing XBP1s can induce the expression of ATF4 and other osteogenic differentiation-related genes and therefore drive osteoblast differentiation. MicroCT analysis further demonstrated that inhibition of XBP1s can strikingly abolish bortezomib-induced bone formation in mouse. CONCLUSIONS These results demonstrated that XBP1s is a master regulator of PI-induced osteoblast differentiation. Activation of IRE1α-XBP1s ER stress signaling can promote osteogenesis, thus providing a novel strategy for the treatment of myeloma bone disease.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Rong Fan
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China.,Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Qiang Jian
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China
| | - Yuchen Zhang
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Lei
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China
| | - Holly Evans
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Yanmeng Wang
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China.,Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Lei Lei
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China
| | - Baiyan Wang
- Department of Clinical Hematology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ramone A Williamson
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China
| | - Andrew Chantry
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Pengcheng He
- Department of Clinical Hematology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Hendrik De Raeve
- Department of Pathology, UZ Brussel, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Aili He
- Department of Clinical Hematology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jinsong Hu
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
31
|
PERK controls bone homeostasis through the regulation of osteoclast differentiation and function. Cell Death Dis 2020; 11:847. [PMID: 33051453 PMCID: PMC7554039 DOI: 10.1038/s41419-020-03046-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022]
Abstract
Osteoclasts are multinucleated giant cells with the ability to degrade bone tissue, and are closely related to abnormal bone metabolic diseases. Endoplasmic reticulum (ER) is an organelle responsible for protein modification, quality control, and transportation. The accumulation of unfolded or misfolded proteins in ER cavity induces ER stress. Double-stranded RNA-dependent protein kinase-like ER kinase (PERK) is an ER stress-sensing protein, which is ubiquitous in eukaryotic cells. Systemic PERK knockout mice show severe bone loss, suggesting that PERK is of great significance for maintaining the normal growth and development of bone tissue, but the role of PERK in osteoclastogenesis is still unclear. In this study, we found that PERK was significantly activated during RANKL-induced osteoclast differentiation; knockdown of PERK by siRNA and inhibition of PERK by GSK2606414, respectively, had significant negative regulatory effects on the formation and bone resorption of osteoclasts. PERK inhibitor GSK2606414 down-regulated the mRNA levels and protein expression of osteoclast differentiation marker genes, and inhibited RANKL-induced activation of Mitogen-activated protein kinase (MAPK) and nuclear factor κB (NF-κB) pathways. Treatment with PERK inhibitor GSK2606414 in ovariectomized mouse model significantly suppressed bone loss and osteoclast formation. Thapsigargin activated ER stress to enhance autophagy, while GSK2606414 had a significant inhibitory effect on autophagy flux and autophagosome formation. Antioxidant N-acetylcysteine (NAC) could inhibit the expression of PERK phosphorylation, osteoclast-related proteins and autophagy-related proteins, but the use of PERK activator CCT020312 can reverse inhibition effect of NAC. Our findings demonstrate a key role for PERK in osteoclast differentiation and suggest its therapeutic potential.
Collapse
|
32
|
Multiple Myeloma-Derived Extracellular Vesicles Induce Osteoclastogenesis through the Activation of the XBP1/IRE1α Axis. Cancers (Basel) 2020; 12:cancers12082167. [PMID: 32759820 PMCID: PMC7465175 DOI: 10.3390/cancers12082167] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Bone disease severely affects the quality of life of over 70% of multiple myeloma (MM) patients, which daily experience pain, pathological fractures, mobility issues and an increased mortality. Recent data have highlighted the crucial role of the endoplasmic reticulum-associated unfolded protein response (UPR) in malignant transformation and tumor progression; therefore, targeting of UPR-related molecules may open novel therapeutic avenues. Endoplasmic reticulum (ER) stress and UPR pathways are constitutively activated in MM cells, which are characterized by an increased protein turnover as a consequence of high production of immunoglobulins and high rates of protein synthesis. A great deal of scientific data also evidenced that a mild activation of UPR pathway can regulate cellular differentiation. Our previous studies revealed that MM cell-derived small extracellular vesicle (MM-EV) modulated osteoclasts (OCs) function and induced OCs differentiation. Here, we investigated the role of the UPR pathway, and in particular of the IRE1α/XBP1 axis, in osteoclastogenesis induced by MM-EVs. By proteomic analysis, we identified UPR signaling molecules as novel MM-EV cargo, prompting us to evaluate the effects of the MM-EVs on osteoclastogenesis through UPR pathway. MM-EVs administration in a murine macrophage cell line rapidly induced activation of IRE1α by phosphorylation in S724; accordingly, Xbp1 mRNA splicing was increased and the transcription of NFATc1, a master transcription factor for OCs differentiation, was activated. Some of these results were also validated using both human primary OC cultures and MM-EVs from MM patients. Notably, a chemical inhibitor of IRE1α (GSK2850163) counteracted MM-EV-triggered OC differentiation, hampering the terminal stages of OCs differentiation and reducing bone resorption.
Collapse
|
33
|
Zhu Y, Sun Y, Zhou Y, Zhang Y, Zhang T, Li Y, You W, Chang X, Yuan L, Han X. MicroRNA-24 promotes pancreatic beta cells toward dedifferentiation to avoid endoplasmic reticulum stress-induced apoptosis. J Mol Cell Biol 2020; 11:747-760. [PMID: 30753517 PMCID: PMC6821228 DOI: 10.1093/jmcb/mjz004] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/02/2018] [Accepted: 02/06/2019] [Indexed: 12/23/2022] Open
Abstract
Current research indicates that beta cell loss in type 2 diabetes may be attributed to beta cell dedifferentiation rather than apoptosis; however, the mechanisms by which this occurs remain poorly understood. Our previous study demonstrated that elevation of microRNA-24 (miR-24) in a diabetic setting caused beta cell dysfunction and replicative deficiency. In this study, we focused on the role of miR-24 in beta cell apoptosis and dedifferentiation under endoplasmic reticulum (ER) stress conditions. We found that miR-24 overabundance protected beta cells from thapsigargin-induced apoptosis at the cost of accelerating the impairment of glucose-stimulated insulin secretion (GSIS) and enhancing the presence of dedifferentiation markers. Ingenuity® Pathway Analysis (IPA) revealed that elevation of miR-24 had an inhibitory effect on XBP1 and ATF4, which are downstream effectors of two key branches of ER stress, by inhibiting its direct target, Ire1α. Notably, elevated miR-24 initiated another pathway that targeted Mafa and decreased GSIS function in surviving beta cells, thus guiding their dedifferentiation under ER stress conditions. Our results demonstrated that the elevated miR-24, to the utmost extent, preserves beta cell mass by inhibiting apoptosis and inducing dedifferentiation. This study not only provides a novel mechanism by which miR-24 dominates beta cell turnover under persistent metabolic stress but also offers a therapeutic consideration for treating diabetes by inducing dedifferentiated beta cells to re-differentiation.
Collapse
Affiliation(s)
- Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Yi Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Yuncai Zhou
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Yan Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Tao Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Weiyan You
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Li Yuan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
34
|
Riaz TA, Junjappa RP, Handigund M, Ferdous J, Kim HR, Chae HJ. Role of Endoplasmic Reticulum Stress Sensor IRE1α in Cellular Physiology, Calcium, ROS Signaling, and Metaflammation. Cells 2020; 9:E1160. [PMID: 32397116 PMCID: PMC7290600 DOI: 10.3390/cells9051160] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) is the most prominent and evolutionarily conserved unfolded protein response (UPR) signal transducer during endoplasmic reticulum functional upset (ER stress). A IRE1α signal pathway arbitrates yin and yang of cellular fate in objectionable conditions. It plays several roles in fundamental cellular physiology as well as in several pathological conditions such as diabetes, obesity, inflammation, cancer, neurodegeneration, and in many other diseases. Thus, further understanding of its molecular structure and mechanism of action during different cell insults helps in designing and developing better therapeutic strategies for the above-mentioned chronic diseases. In this review, recent insights into structure and mechanism of activation of IRE1α along with its complex regulating network were discussed in relation to their basic cellular physiological function. Addressing different binding partners that can modulate IRE1α function, UPRosome triggers different downstream pathways depending on the cellular backdrop. Furthermore, IRE1α are in normal cell activities outside the dominion of ER stress and activities under the weather of inflammation, diabetes, and obesity-related metaflammation. Thus, IRE1 as an ER stress sensor needs to be understood from a wider perspective for comprehensive functional meaning, which facilitates us with assembling future needs and therapeutic benefits.
Collapse
Affiliation(s)
- Thoufiqul Alam Riaz
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| | - Raghu Patil Junjappa
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| | - Mallikarjun Handigund
- Department of Laboratory Medicine, Jeonbuk National University, Medical School, Jeonju 54907, Korea;
| | - Jannatul Ferdous
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea;
| | - Hyung-Ryong Kim
- College of Dentistry, Dankook University, Cheonan 31116, Korea
| | - Han-Jung Chae
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| |
Collapse
|
35
|
Iyer S, Melendez-Suchi C, Han L, Baldini G, Almeida M, Jilka RL. Elevation of the unfolded protein response increases RANKL expression. FASEB Bioadv 2020; 2:207-218. [PMID: 32259048 PMCID: PMC7133738 DOI: 10.1096/fba.2019-00032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/12/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
Increased production of the osteoclastogenic cytokine RANKL is a common feature of pathologic bone loss, but the underlying cause of this increase is poorly understood. The unfolded protein response (UPR) is activated in response to accumulation of misfolded proteins in the endoplasmic reticulum (ER). Failure to resolve misfolding results in excess UPR signaling that stimulates cytokine production and cell death. We therefore investigated whether RANKL is one of the cytokines stimulated in response to elevated UPR in bone cells. Pharmacologic induction of UPR with tunicamycin (Tm)-stimulated RANKL expression in cultures of primary osteoblastic cells and in osteoblast and osteocyte cell lines. Pharmacologic inhibition of the UPR blunted Tm-induced RANKL production. Silencing Edem1 or Sel1l, proteins that aid in degradation of misfolded proteins, also induced UPR and increased RANKL mRNA. Moreover, Tm or hypoxia increased RANKL and bone resorption in cultures of neonatal murine calvaria. Administration of Tm to adult mice caused dilation of ER in osteoblasts and osteocytes, elevated the UPR, and increased RANKL expression and osteoclast number. These findings support the hypothesis that excessive UPR signaling stimulates the expression of RANKL by osteoblasts and osteocytes, and thereby facilitates excessive bone resorption and bone loss in pathologic conditions.
Collapse
Affiliation(s)
- Srividhya Iyer
- Department of Orthopaedic Surgery University of Arkansas Medical Sciences Little Rock AR USA
| | | | - Li Han
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology University of Arkansas Medical Sciences Little Rock AR USA
| | - Maria Almeida
- Department of Orthopaedic Surgery University of Arkansas Medical Sciences Little Rock AR USA
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
| | - Robert L Jilka
- Division of Endocrinology and Metabolism Center for Osteoporosis and Metabolic Bone Diseases University of Arkansas Medical Sciences Little Rock AR USA
- Central Arkansas Veterans Healthcare System Little Rock AR USA
| |
Collapse
|
36
|
Ma Q, Liang M, Wang Y, Ding N, Wu Y, Duan L, Yu T, Lu Y, Xu J, Kang F, Dou C. Non-coenzyme role of vitamin B1 in RANKL-induced osteoclastogenesis and ovariectomy induced osteoporosis. J Cell Biochem 2020; 121:3526-3536. [PMID: 32100911 DOI: 10.1002/jcb.29632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/09/2019] [Indexed: 01/29/2023]
Abstract
Vitamins B are co-enzymes participating in energy metabolic pathways. While some vitamins B are known affecting bone homeostasis, the effects of vitamin B1 (thiamine) on bone health remains unclear. In our study, we used cell counting kit-8, tartrate-resistant acid phosphatase stain, actin cytoskeleton stain, and pit formation assay to evaluate the effect of thiamine on osteoclast differentiation, formation, and function, respectively. Then we used dichloro-dihydro-fluorescein diacetate assay to investigate reactive oxygen species (ROS) generation and removal. Osteoporosis model by ovariectomy was established for animal experiments. We found that thiamine had inhibitory effect on osteoclast differentiation. And its inhibitory role on osteoclast differentiation is in a dose-dependent way. Mechanistically, ThDP suppresses intracellular ROS accumulation and unfolded protein response signaling during osteoclastogenesis via inhibiting Rac-Nox1/2/4 and intracellular inositol-requiring protein-1α/X-box-binding protein pathways, respectively. Osteoporotic mice treated with thiamine rich dietary showed better bone strength relative to thiamine deficient dietary. Our study explored the non-coenzyme inhibitory functions of B1 vitamin in receptor activator of nuclear factor κB ligand induced osteoclastogenesis and uncovered the significance of B1 vitamin in bone health.
Collapse
Affiliation(s)
- Qinyu Ma
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Mengmeng Liang
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Yaxi Wang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ning Ding
- Department of Blood Purification, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Yutong Wu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lianli Duan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanzhu Lu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Fei Kang
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
37
|
Li N, Inoue K, Sun J, Niu Y, Lalani S, Yallowitz A, Yang X, Zhang C, Shen R, Zhao B, Xu R, Greenblatt MB. Osteoclasts are not a source of SLIT3. Bone Res 2020; 8:11. [PMID: 32133214 PMCID: PMC7031526 DOI: 10.1038/s41413-020-0086-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 09/29/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022] Open
Abstract
The axon guidance cue SLIT3 was identified as an osteoanabolic agent in two recent reports. However, these reports conflict in their nomination of osteoblasts versus osteoclasts as the key producers of skeletal SLIT3 and additionally offer conflicting data on the effects of SLIT3 on osteoclastogenesis. Here, aiming to address this discrepancy, we found no observable SLIT3 expression during human or mouse osteoclastogenesis and the only modest SLIT3-mediated effects on osteoclast differentiation. Conditional deletion of SLIT3 in cathepsin K (CTSK)-positive cells, including osteoclasts, had no effect on the number of osteoclast progenitors, in vitro osteoclast differentiation, overall bone mass, or bone resorption/formation parameters. Similar results were observed with the deletion of SLIT3 in LysM-positive cells, including osteoclast lineage cells. Consistent with this finding, bone marrow chimeras made from Slit3 -/- donors that lacked SLIT3 expression at all stages of osteoclast development displayed normal bone mass relative to controls. Taken in context, multiple lines of evidence were unable to identify the physiologic function of osteoclast-derived SLIT3, indicating that osteoblasts are the major source of skeletal SLIT3.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian China
| | - Kazuki Inoue
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY USA
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY USA
| | - Yingzhen Niu
- Division of Adult Reconstruction and Joint Replacement, Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY USA
| | - Sarfaraz Lalani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY USA
| | - Alisha Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY USA
| | - Xu Yang
- Division of Adult Reconstruction and Joint Replacement, Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY USA
| | - Chao Zhang
- Institute for Computational Biomedicine, Cornell University, New York, NY USA
| | - Rong Shen
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian China
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY USA
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, Fujian China
| | - Matthew B. Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY USA
| |
Collapse
|
38
|
Chang Y, Yu D, Chu W, Liu Z, Li H, Zhai Z. LncRNA expression profiles and the negative regulation of lncRNA-NOMMUT037835.2 in osteoclastogenesis. Bone 2020; 130:115072. [PMID: 31593824 DOI: 10.1016/j.bone.2019.115072] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 09/10/2019] [Accepted: 09/14/2019] [Indexed: 12/19/2022]
Abstract
Bone is a rigid and dynamic organ that continuously undergoes remodeling and repair. The balance between osteoblastic bone formation and osteoclastic bone resorption is essential for normal bone homeostasis. Osteoclasts are giant multinucleated cells derived from the monocyte/macrophage hematopoietic lineage and are regulated by various cytokines. Long non-coding (lnc) RNAs are known to regulate many biological processes in the skeletal system in both normal and diseased states; however, the lncRNA-mediated regulation of osteoclastogenesis has not been extensively studied. Hence, in the present study, we performed microarray analysis of lncRNAs expressed during different stages of osteoclast differentiation and fusion. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed the biological functions of target genes of lncRNAs that were specifically up- or downregulated at the different stages. Microarray and bioinformatic prediction results were used to generate co-expression networks of lncRNAs-mRNAs and lncRNAs-transcription factors. Based on the analysis, we identified one lncRNA, NONMMUT037835.2, which plays an important role during osteoclastogenesis. Upregulation of lncRNA-NONMMUT037835.2 inhibited osteoclastic differentiation, whereas downregulation of lncRNA-NONMMUT037835.2 promoted osteoclast formation and fusion. Our study also indicated that lncRNA-NOMMUT037835.2 might regulated osteoclastogenesis through negatively regulating RANK expression and inhibiting NF-κB/MAPK signaling pathway. Our results lead to a better understanding of the molecular mechanisms and provided a theoretical basis for developing therapeutic agents for diseases related to dysregulation of bone homeostasis.
Collapse
Affiliation(s)
- Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Degang Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wenxiang Chu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhiqing Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
39
|
Yoon SB, Park YH, Choi SA, Yang HJ, Jeong PS, Cha JJ, Lee S, Lee SH, Lee JH, Sim BW, Koo BS, Park SJ, Lee Y, Kim YH, Hong JJ, Kim JS, Jin YB, Huh JW, Lee SR, Song BS, Kim SU. Real-time PCR quantification of spliced X-box binding protein 1 (XBP1) using a universal primer method. PLoS One 2019; 14:e0219978. [PMID: 31329612 PMCID: PMC6645673 DOI: 10.1371/journal.pone.0219978] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/05/2019] [Indexed: 12/21/2022] Open
Abstract
X-box binding protein 1 (XBP1) mRNA processing plays a crucial role in the unfolded protein response (UPR), which is activated in response to endoplasmic reticulum (ER) stress. Upon accumulation of the UPR-converted XBP1 mRNA splicing from an unspliced (u) XBP1 (inactive) isoform to the spliced (s) XBP1 (active) isoform, inositol-requiring enzyme 1 α (IRE1α) removes a 26-nucleotide intron from uXBP1 mRNA. Recent studies have reported the assessment of ER stress by examining the ratio of sXBP1 to uXBP1 mRNA (s/uXBP1 ratio) via densitometric analysis of PCR bands relative to increased levels of sXBP1 to uXBP1 using a housekeeping gene for normalization. However, this measurement is visualized by gel electrophoresis, making it very difficult to quantify differences between the two XBP1 bands and complicating data interpretation. Moreover, most commonly used housekeeping genes display an unacceptably high variable expression pattern of the s/uXBP1 ratio under different experimental conditions, such as various phases of development and different cell types, limiting their use as internal controls. For a more quantitative determination of XBP1 splicing activity, we measured the expression levels of total XBP1 (tXBP1: common region of s/uXBP1) and sXBP1 via real-time PCR using specific primer sets. We also designed universal real-time PCR primer sets capable of amplifying a portion of each u/s/tXBP1 mRNA that is highly conserved in eukaryotes, including humans, monkeys, cows, pigs, and mice. Therefore, we provide a more convenient and easily approachable quantitative real-time PCR method that can be used in various research fields to assess ER stress.
Collapse
Affiliation(s)
- Seung-Bin Yoon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Primate Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeollabuk-do, Republic of Korea
| | - Young-Ho Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Seon-A Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Hae-Jun Yang
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Pil-Soo Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Jae-Jin Cha
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Sanghoon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Seung Hwan Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Jong-Hee Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Bo-Woong Sim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Bon-Sang Koo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Sang-Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Ji-Su Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Primate Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeollabuk-do, Republic of Korea
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Bong-Seok Song
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
- * E-mail: (BSS); (SUK)
| | - Sun-Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
- * E-mail: (BSS); (SUK)
| |
Collapse
|
40
|
Bezamat M, Deeley K, Khaliq S, Letra A, Scariot R, Silva RM, Weber ML, Bussaneli DG, Trevilatto PC, Almarza AJ, Ouyang H, Vieira AR. Are mTOR and Endoplasmic Reticulum Stress Pathway Genes Associated with Oral and Bone Diseases? Caries Res 2018; 53:235-241. [PMID: 30205378 DOI: 10.1159/000492675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
The purpose of this cohort study was to identify associations between combined oral and bone disease phenotypes and genes present in cell regulatory pathways. The studied pathways play important roles in cellular growth, proliferation, differentiation, and homeostasis. DNA samples extracted from whole saliva of 3,912 individuals were genotyped and these data analyzed according to dental caries experience, periapical lesions, periodontitis, osteoporosis, or temporomandibular joint discomfort. Samples were obtained from the Dental Registry and DNA Repository project at the University of Pittsburgh. Twenty-seven polymorphisms in eight genes related to mTOR or endoplasmic reticulum stress pathways were selected for genotyping. Allele frequencies and Hardy-Weinberg equilibrium were calculated. Analyses were performed comparing genotypes between affected and unaffected individuals for each phenotype, as well as for the associated phenotypes combined. For all analyses, we used the software PLINK with an alpha of 0.002. Borderline associations with multiple variants of several genes were found, suggesting that both pathways may be involved in the susceptibility to multiple conditions affecting the oral cavity and bones. When combining patients that had concomitant dental caries, periodontitis, and periapical pathology, several markers in RHEB showed statistically significant association. Multiple conditions affecting bone and teeth (i.e., dental caries, periodontitis, periapical lesion formation, and osteoporosis) appear to share similar underlying genetic etiological factors, which allow us to hypothesize that instead of individually, they should be studied in conjunction in human populations.
Collapse
Affiliation(s)
- Mariana Bezamat
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kathleen Deeley
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shahryar Khaliq
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ariadne Letra
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Endodontics, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Rafaela Scariot
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Graduate Program of Health Sciences, PUC-Paraná, Curitiba, Brazil.,Department of Oral Surgery, Positivo University, Curitiba, Brazil
| | - Renato M Silva
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Endodontics, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Megan L Weber
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Diego G Bussaneli
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pediatric Dentistry, UNESP, Araraquara, Brazil
| | | | - Alejandro J Almarza
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hongjiao Ouyang
- Department of Endodontics, Texas A&M College of Dentistry, Dallas, Texas, USA
| | - Alexandre R Vieira
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA, .,Graduate Program of Health Sciences, PUC-Paraná, Curitiba, Brazil,
| |
Collapse
|
41
|
Junjappa RP, Patil P, Bhattarai KR, Kim HR, Chae HJ. IRE1α Implications in Endoplasmic Reticulum Stress-Mediated Development and Pathogenesis of Autoimmune Diseases. Front Immunol 2018; 9:1289. [PMID: 29928282 PMCID: PMC5997832 DOI: 10.3389/fimmu.2018.01289] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022] Open
Abstract
Inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) is the most prominent and evolutionarily conserved endoplasmic reticulum (ER) membrane protein. This transduces the signal of misfolded protein accumulation in the ER, named as ER stress, to the nucleus as “unfolded protein response (UPR).” The ER stress-mediated IRE1α signaling pathway arbitrates the yin and yang of cell life. IRE1α has been implicated in several physiological as well as pathological conditions, including immune disorders. Autoimmune diseases are caused by abnormal immune responses that develop due to genetic mutations and several environmental factors, including infections and chemicals. These factors dysregulate the cell immune reactions, such as cytokine secretion, antigen presentation, and autoantigen generation. However, the mechanisms involved, in which these factors induce the onset of autoimmune diseases, are remaining unknown. Considering that these environmental factors also induce the UPR, which is expected to have significant role in secretory cells and immune cells. The role of the major UPR molecule, IRE1α, in causing immune responses is well identified, but its role in inducing autoimmunity and the pathogenesis of autoimmune diseases has not been clearly elucidated. Hence, a better understanding of the role of IRE1α and its regulatory mechanisms in causing autoimmune diseases could help to identify and develop the appropriate therapeutic strategies. In this review, we mainly center the discussion on the molecular mechanisms of IRE1α in the pathophysiology of autoimmune diseases.
Collapse
Affiliation(s)
- Raghu Patil Junjappa
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Prakash Patil
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Kashi Raj Bhattarai
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| | - Hyung-Ryong Kim
- Graduate School, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | - Han-Jung Chae
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Chonbuk National University, Jeonju, South Korea
| |
Collapse
|
42
|
Sylvester FA. Inflammatory Bowel Disease: Effects on Bone and Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1033:133-150. [PMID: 29101654 DOI: 10.1007/978-3-319-66653-2_7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD) is associated with decreased bone mass and alterations in bone geometry from the time of diagnosis, before anti-inflammatory therapy is instituted. Deficits in bone mass can persist despite absence of symptoms of active IBD. The effects of IBD on the skeleton are complex. Protein-calorie malnutrition, inactivity, hypogonadism, deficits in calcium intake and vitamin D consumption and synthesis, stunted growth in children, decreased skeletal muscle mass, and inflammation all likely play a role. Preliminary studies suggest that the dysbiotic intestinal microbial flora present in IBD may also affect bone at a distance. Several mechanisms are possible. T cells activated by the gut microbiota may serve as "inflammatory shuttles" between the intestine and bone. Microbe-associated molecular patterns leaked into the circulation in IBD may activate immune responses in the bone marrow by immune cells and by osteocytes, osteoblasts, and osteoclasts that lead to decreased bone formation and increased resorption. Finally, intestinal microbial metabolites such as H2S may also affect bone cell function. Uncovering these mechanisms will enable the design of microbial cocktails to help restore bone mass in patients with IBD.
Collapse
Affiliation(s)
- Francisco A Sylvester
- Division Chief of Pediatric Gastroenterology, The University of North Carolina at Chapel Hil, 333 South Columbia Street, MacNider Hall 247, Chapel Hill, NC, 27599-7229, USA.
| |
Collapse
|
43
|
Ding N, Liu C, Yao L, Bai Y, Cheng P, Li Z, Luo K, Mei T, Li J, Xing J, Gao X, Ma Q, Xu J, Luo F, Dou C. Alendronate induces osteoclast precursor apoptosis via peroxisomal dysfunction mediated ER stress. J Cell Physiol 2018; 233:7415-7423. [PMID: 29600563 DOI: 10.1002/jcp.26587] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/08/2018] [Indexed: 12/23/2022]
Abstract
Nitrogen-containing bisphosphonates including alendronate (ALN) are the current first line antiresorptive drug in treating osteoporosis. In our study, we found that ALN administration impaired the secretion of platelet derived growth factor-BB (PDGF-BB), the most important angiogenic cytokines produced by preosteoclast (POC), in both sham and ovariectomized (OVX) mice. To further understand this phenomenon, we induced bone marrow macrophages (BMMs) to POCs in vitro and detected the effects of ALN particularly in POCs. The proapoptotic effect of ALN in POCs was confirmed by flow cytometry. On the molecular level, we found that farnesyl diphosphate synthase (FDPS) inhibition of ALN led to peroxisomal dysfunction and up regulation of cytoprotective protein glucose-regulated protein (GRP) 78. Peroxisomal dysfunction further induced endoplasmic reticulum (ER) stress in POCs and finally resulted in cell apoptosis marked by reduced expression of B-cell lymphoma 2 (Bcl-2) and increased expressions of CCAAT/enhancer binding protein homologous protein (CHOP), Bcl2 associated X (Bax), and cleaved caspase-3. We concluded that ALN has no selectivity in inhibiting POC and mature osteoclast. For POCs, ALN inhibition of FDPS leads to peroxisomal dysfunction, which further mediates ER stress and finally causes cell apoptosis. Considering that decreased angiogenesis is also an important issue in treating osteoporosis, how to preserve pro-angiogenic POCs while depleting mature osteoclasts is a problem worthy to be solved.
Collapse
Affiliation(s)
- Ning Ding
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuan Liu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Urology, The Army General Hospital, Beijing, China
| | - Li Yao
- Department of Urology, The Army General Hospital, Beijing, China
| | - Yun Bai
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Peng Cheng
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhilin Li
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Keyu Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tieniu Mei
- Department of Surgery, Shigatse Branch of Xinqiao Hospital, The Third Military Medical University (Army Medical University), Shigatse, China
| | - Jianhua Li
- Department of Orthopedics, The 88 Hospital of PLA, Taian, China
| | - Junchao Xing
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoliang Gao
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qinyu Ma
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
44
|
Iseme RA, Mcevoy M, Kelly B, Agnew L, Walker FR, Attia J. Is osteoporosis an autoimmune mediated disorder? Bone Rep 2017; 7:121-131. [PMID: 29124082 PMCID: PMC5671387 DOI: 10.1016/j.bonr.2017.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 08/01/2017] [Accepted: 10/15/2017] [Indexed: 12/12/2022] Open
Abstract
The last two decades have marked a growing understanding of the interaction occurring between bone and immune cells. The chronic inflammation and immune system dysfunction commonly observed to occur during the ageing process and as part of a range of other pathological conditions, commonly associated with osteoporosis has led to the recognition of these processes as important determinants of bone disease. This is further supported by the recognition that the immune and bone systems in fact share regulatory mechanisms and progenitor molecules. Research into this complex synergy has provided a better understanding of the immunopathogenesis underlying bone diseases such as osteoporosis. However, existing research has largely focussed on delineating the role played by inflammation in pathogenic bone destruction, despite increasing evidence implicating autoantibodies as important drivers of osteoporosis. This review shall attempt to provide a comprehensive overview of existing research examining the role played by autoantibodies in osteoporosis in order to determine the potential for further research in this area. Autoantibodies represent promising targets for the improved treatment and diagnosis of inflammatory bone loss.
Collapse
Affiliation(s)
- Rosebella A. Iseme
- Department of Population and Reproductive Health, School of Public Health, Kenyatta University, P.O. Box 43844 –, 00100, Nairobi, Kenya
- School of Medicine & Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Mark Mcevoy
- School of Medicine & Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
| | - Brian Kelly
- School of Medicine & Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Centre for Brain and Mental Health Research, The University of Newcastle, Callaghan, NSW, Australia
| | - Linda Agnew
- Brain Behaviour Research Group, School of Science and Technology, University of New England, Armidale, NSW 2351, Australia
| | - Frederick R. Walker
- School of Medicine & Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Laboratory of Affective Neuroscience, The University of Newcastle, Callaghan, NSW, Australia
- University of Newcastle, Medical Sciences MS413, University Drive, Callaghan, NSW 2308, Australia
| | - John Attia
- School of Medicine & Public Health, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW, 2305, Australia
- Department of General Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| |
Collapse
|
45
|
Kaufman DR, Papillon J, Larose L, Iwawaki T, Cybulsky AV. Deletion of inositol-requiring enzyme-1α in podocytes disrupts glomerular capillary integrity and autophagy. Mol Biol Cell 2017; 28:1636-1651. [PMID: 28428258 PMCID: PMC5469607 DOI: 10.1091/mbc.e16-12-0828] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/10/2017] [Accepted: 04/14/2017] [Indexed: 12/02/2022] Open
Abstract
Inositol-requiring enzyme-1α (IRE1α) is an endoplasmic reticulum (ER)-transmembrane endoribonuclease kinase that plays an essential function in extraembryonic tissues during normal development and is activated during ER stress. To address the functional role of IRE1α in glomerular podocytes, we produced podocyte-specific IRE1α-deletion mice. In male mice, deletion of IRE1α in podocytes resulted in albuminuria beginning at 5 mo of age and worsening with time. Electron microscopy revealed focal podocyte foot-process effacement in 9-mo-old male IRE1α-deletion mice, as well as microvillous transformation of podocyte plasma membranes. Compared with control, glomerular cross-sectional and capillary lumenal areas were greater in deletion mice, and there was relative podocyte depletion. Levels of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II expression and c-Jun N-terminal kinase-1 phosphorylation were decreased in IRE1α-deletion glomeruli, in keeping with reduced autophagy. Deletion of IRE1α exacerbated glomerular injury in anti-glomerular basement membrane nephritis. In cell culture, IRE1α dominant-negative mutants reduced the physiological (basal) accumulation of LC3B-II and the size of autophagic vacuoles but did not affect ER-associated degradation. Thus IRE1α is essential for maintaining podocyte and glomerular integrity as mice age and in glomerulonephritis. The mechanism is related, at least in part, to the maintenance of autophagy in podocytes.
Collapse
Affiliation(s)
- Daniel Robert Kaufman
- Department of Physiology, McGill University, and McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| | - Joan Papillon
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| | - Louise Larose
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| | - Takao Iwawaki
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Andrey V Cybulsky
- Department of Physiology, McGill University, and McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
46
|
Shimoda M, Yoshida H, Mizuno S, Hirozane T, Horiuchi K, Yoshino Y, Hara H, Kanai Y, Inoue S, Ishijima M, Okada Y. Hyaluronan-Binding Protein Involved in Hyaluronan Depolymerization Controls Endochondral Ossification through Hyaluronan Metabolism. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1162-1176. [PMID: 28284715 DOI: 10.1016/j.ajpath.2017.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/25/2016] [Accepted: 01/11/2017] [Indexed: 11/18/2022]
Abstract
Hyaluronan (HA) plays an important role in the development and maintenance of tissues, and its degradation is implicated in many pathologic conditions. We recently reported that HA-binding protein involved in HA depolymerization (CEMIP; alias HYBID/KIAA1199) is a key molecule in HA depolymerization, but its developmental and pathologic functions remain elusive. We generated Hybid-deficient mice using the Cre/locus of crossover in P1 (loxP) system and analyzed their phenotypes. Hybid-deficient mice were viable and fertile, but their adult long bones were shorter than those of wild-type animals. Hybid-deficient mice showed lengthening of hypertrophic zone in the growth plate until 4 weeks after birth. There were fewer capillaries and osteoclasts at the chondroosseous junction in the Hybid-deficient mice compared with the wild-type mice. In situ hybridization demonstrated that Hybid was expressed by hypertrophic chondrocytes at the chondroosseous junction. Cultured primary chondrocytes expressed higher levels of Hybid than did osteoblasts or osteoclasts, and the Hybid expression in the chondrocytes was up-regulated after maturation to hypertrophic chondrocytes. High-molecular-weight HA was accumulated in the lengthened hypertrophic zone in Hybid-deficient mice. In addition, high-molecular-weight HA significantly reduced cell growth and tube formation in vascular endothelial growth factor-stimulated or -nonstimulated endothelial cells. HA metabolism by HYBID is involved in endochondral ossification during postnatal development by modulation of angiogenesis and osteoclast recruitment at the chondroosseous junction.
Collapse
Affiliation(s)
- Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.
| | | | - Sakiko Mizuno
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Toru Hirozane
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Keisuke Horiuchi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Yoshino
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Inoue
- Cosmetic Health Science, Gifu Pharmaceutical University, Gifu, Japan
| | - Muneaki Ishijima
- Department of Orthopaedic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan; Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
47
|
Inhibition of IRE1α-driven pro-survival pathways is a promising therapeutic application in acute myeloid leukemia. Oncotarget 2017; 7:18736-49. [PMID: 26934650 PMCID: PMC4951325 DOI: 10.18632/oncotarget.7702] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/29/2016] [Indexed: 01/07/2023] Open
Abstract
Survival of cancer cells relies on the unfolded protein response (UPR) to resist stress triggered by the accumulation of misfolded proteins within the endoplasmic reticulum (ER). The IRE1α-XBP1 pathway, a key branch of the UPR, is activated in many cancers. Here, we show that the expression of both mature and spliced forms of XBP1 (XBP1s) is up-regulated in acute myeloid leukemia (AML) cell lines and AML patient samples. IRE1α RNase inhibitors [MKC-3946, 2-hydroxy-1-naphthaldehyde (HNA), STF-083010 and toyocamycin] blocked XBP1 mRNA splicing and exhibited cytotoxicity against AML cells. IRE1α inhibition induced caspase-dependent apoptosis and G1 cell cycle arrest at least partially by regulation of Bcl-2 family proteins, G1 phase controlling proteins (p21cip1, p27kip1 and cyclin D1), as well as chaperone proteins. Xbp1 deleted murine bone marrow cells were resistant to growth inhibition by IRE1α inhibitors. Combination of HNA with either bortezomib or AS2O3 was synergistic in AML cytotoxicity associated with induction of p-JNK and reduction of p-PI3K and p-MAPK. Inhibition of IRE1α RNase activity increased expression of many miRs in AML cells including miR-34a. Inhibition of miR-34a conferred cellular resistance to HNA. Our results strongly suggest that targeting IRE1α driven pro-survival pathways represent an exciting therapeutic approach for the treatment of AML.
Collapse
|
48
|
Navid F, Colbert RA. Causes and consequences of endoplasmic reticulum stress in rheumatic disease. Nat Rev Rheumatol 2016; 13:25-40. [PMID: 27904144 DOI: 10.1038/nrrheum.2016.192] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Rheumatic diseases represent a heterogeneous group of inflammatory conditions, many of which involve chronic activation of both innate and adaptive immune responses by multiple genetic and environmental factors. These immune responses involve the secretion of excessive amounts of cytokines and other signalling mediators by activated immune cells. The endoplasmic reticulum (ER) is the cellular organelle that directs the folding, processing and trafficking of membrane-bound and secreted proteins, including many key components of the immune response. Maintaining homeostasis in the ER is critical to cell function and survival. Consequently, elaborate mechanisms have evolved to sense and respond to ER stress through three main signalling pathways that together comprise the unfolded protein response (UPR). Activation of the UPR can rapidly resolve the accumulation of misfolded proteins, direct permanent changes in the size and function of cells during differentiation, and critically influence the immune response and inflammation. Recognition of the importance of ER stress and UPR signalling pathways in normal and dysregulated immune responses has greatly increased in the past few years. This Review discusses several settings in which ER stress contributes to the pathogenesis of rheumatic diseases and considers some of the therapeutic opportunities that these discoveries provide.
Collapse
Affiliation(s)
- Fatemeh Navid
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Building 10, Room 12N248B,10 Center Drive, Bethesda, Maryland 20892, USA
| | - Robert A Colbert
- Pediatric Translational Research Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Building 10, Room 12N248B,10 Center Drive, Bethesda, Maryland 20892, USA
| |
Collapse
|
49
|
Hirozane T, Tohmonda T, Yoda M, Shimoda M, Kanai Y, Matsumoto M, Morioka H, Nakamura M, Horiuchi K. Conditional abrogation of Atm in osteoclasts extends osteoclast lifespan and results in reduced bone mass. Sci Rep 2016; 6:34426. [PMID: 27677594 PMCID: PMC5039636 DOI: 10.1038/srep34426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 09/13/2016] [Indexed: 01/06/2023] Open
Abstract
Ataxia-telangiectasia mutated (ATM) kinase is a central component involved in the signal transduction of the DNA damage response (DDR) and thus plays a critical role in the maintenance of genomic integrity. Although the primary functions of ATM are associated with the DDR, emerging data suggest that ATM has many additional roles that are not directly related to the DDR, including the regulation of oxidative stress signaling, insulin sensitivity, mitochondrial homeostasis, and lymphocyte development. Patients and mice lacking ATM exhibit growth retardation and lower bone mass; however, the mechanisms underlying the skeletal defects are not fully understood. In the present study, we generated mutant mice in which ATM is specifically inactivated in osteoclasts. The mutant mice did not exhibit apparent developmental defects but showed reduced bone mass due to increased osteoclastic bone resorption. Osteoclasts lacking ATM were more resistant to apoptosis and showed a prolonged lifespan compared to the controls. Notably, the inactivation of ATM in osteoclasts resulted in enhanced NF-κB signaling and an increase in the expression of NF-κB-targeted genes. The present study reveals a novel function for ATM in regulating bone metabolism by suppressing the lifespan of osteoclasts and osteoclast-mediated bone resorption.
Collapse
Affiliation(s)
- Toru Hirozane
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
- Japan Society for the Promotion of Science, Tokyo 102-8472, Japan
| | - Takahide Tohmonda
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Anti-Aging Orthopedic Research, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Yoda
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Anti-Aging Orthopedic Research, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hideo Morioka
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Anti-Aging Orthopedic Research, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Horiuchi K, Tohmonda T, Morioka H. The unfolded protein response in skeletal development and homeostasis. Cell Mol Life Sci 2016; 73:2851-69. [PMID: 27002737 PMCID: PMC11108572 DOI: 10.1007/s00018-016-2178-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/06/2016] [Accepted: 03/10/2016] [Indexed: 12/20/2022]
Abstract
Osteoblasts and chondrocytes produce a large number of extracellular matrix proteins to generate and maintain the skeletal system. To cope with their functions as secretory cells, these cells must acquire a considerable capacity for protein synthesis and also the machinery for the quality-control and transport of newly synthesized secreted proteins. The unfolded protein response (UPR) plays a crucial role during the differentiation of these cells to achieve this goal. Unexpectedly, however, studies in the past several years have revealed that the UPR has more extensive functions in skeletal development than was initially assumed, and the UPR critically orchestrates many facets of skeletal development and homeostasis. This review focuses on recent findings on the functions of the UPR in the differentiation of osteoblasts, chondrocytes, and osteoclasts. These findings may have a substantial impact on our understanding of bone metabolism and also on establishing treatments for congenital and acquired skeletal disorders.
Collapse
Affiliation(s)
- Keisuke Horiuchi
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
- Department of Anti-aging Orthopedic Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Takahide Tohmonda
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Anti-aging Orthopedic Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideo Morioka
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|