1
|
Li C, Sun Y, Xu W, Chang F, Wang Y, Ding J. Mesenchymal Stem Cells-Involved Strategies for Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305116. [PMID: 38477559 PMCID: PMC11200100 DOI: 10.1002/advs.202305116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/13/2023] [Indexed: 03/14/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the joints and bone destruction. Because of systemic administration and poor targeting, traditional anti-rheumatic drugs have unsatisfactory treatment efficacy and strong side effects, including myelosuppression, liver or kidney function damage, and malignant tumors. Consequently, mesenchymal stem cells (MSCs)-involved therapy is proposed for RA therapy as a benefit of their immunosuppressive and tissue-repairing effects. This review summarizes the progress of MSCs-involved RA therapy through suppressing inflammation and promoting tissue regeneration and predicts their potential clinical application.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Yifu Sun
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Fei Chang
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Yinan Wang
- Department of BiobankDivision of Clinical ResearchThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of EducationThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
2
|
Liao Z, Zheng X, Li H, Deng Z, Feng S, Tan H, Zhao L. Carboxypeptidase M modulates BMSCs osteogenesis-adipogenesis via the MAPK/ERK pathway: An integrated single-cell and bulk transcriptomic study. FASEB J 2024; 38:e23657. [PMID: 38713087 DOI: 10.1096/fj.202302508r] [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: 12/06/2023] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024]
Abstract
The pathogenesis of osteoporosis (OP) is closely associated with the disrupted balance between osteogenesis and adipogenesis in bone marrow-derived mesenchymal stem cells (BMSCs). We analyzed published single-cell RNA sequencing (scRNA-seq) data to dissect the transcriptomic profiles of bone marrow-derived cells in OP, reviewing 56 377 cells across eight scRNA-seq datasets from femoral heads (osteoporosis or osteopenia n = 5, osteoarthritis n = 3). Seventeen genes, including carboxypeptidase M (CPM), were identified as key osteogenesis-adipogenesis regulators through comprehensive gene set enrichment, differential expression, regulon activity, and pseudotime analyses. In vitro, CPM knockdown reduced osteogenesis and promoted adipogenesis in BMSCs, while adenovirus-mediated CPM overexpression had the reverse effects. In vivo, intraosseous injection of CPM-overexpressing BMSCs mitigated bone loss in ovariectomized mice. Integrated scRNA-seq and bulk RNA sequencing analyses provided insight into the MAPK/ERK pathway's role in the CPM-mediated regulation of BMSC osteogenesis and adipogenesis; specifically, CPM overexpression enhanced MAPK/ERK signaling and osteogenesis. In contrast, the ERK1/2 inhibitor binimetinib negated the effects of CPM overexpression. Overall, our findings identify CPM as a pivotal regulator of BMSC differentiation, which provides new clues for the mechanistic study of OP.
Collapse
Affiliation(s)
- Zheting Liao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoyong Zheng
- Orthopaedic Department, The 4th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongfang Li
- Beijing Yijiandian Clinic, Beijing, China
- Health Management Center, Peking University International Hospital, Beijing, China
| | - Zhonghao Deng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuhao Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongbo Tan
- Department of Orthopaedic, The 920th Hospital of Joint Logistics Support Force, Kunming, Yunnan, China
| | - Liang Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Chen Z, Cao Y, Jiang W, Yan Z, Cai G, Ye J, Wang H, Liu L. Porphyromonas gingivalis OMVs promoting endothelial dysfunction via the STING pathway in periodontitis. Oral Dis 2024. [PMID: 38696515 DOI: 10.1111/odi.14969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 05/04/2024]
Abstract
OBJECTIVE This study aimed to assess the effects of Porphyromonas gingivalis outer membrane vesicles (Pg-OMVs) in chronic periodontitis and explore the underlying mechanism involved. METHODS In vitro, Pg-OMVs were incubated with Ea.hy926 (vessel endothelial cells, ECs) to evaluate their effects on endothelial functions and to investigate the underlying mechanism. The effects of endothelial dysfunction on MG63 osteoblast-like cells were verified using an indirect co-culture method. For in vivo studies, micro-CT was conducted to identify alveolar bone mass. Immunofluorescence staining was conducted to confirm the levels of stimulator of interferon genes (STING) in the blood vessel and the number of Runx2+ cells around the alveolar bone. RESULTS Pg-OMVs were endocytosed by ECs, leading to endothelial dysfunction. The cGAS-STING-TBK1 pathway was activated in ECs, which subsequently inhibited MG63 migration and early osteogenesis differentiation. In vivo, Pg-OMVs promoted alveolar bone resorption, increased STING levels in the blood vessel, and decreased Runx2+ cells around the alveolar bone. CONCLUSIONS Pg-OMVs caused endothelial dysfunction and activated the cGAS-STING-TBK1 signal cascade in ECs, thereby impairing ECs-mediated osteogenesis. Furthermore, Pg-OMVs aggregated alveolar bone loss and altered the blood vessel-mediated osteogenesis with elevated STING.
Collapse
Affiliation(s)
- Zhenwei Chen
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Ye Cao
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Wenxiu Jiang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Zixin Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Guanhui Cai
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Junjie Ye
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
| | - Hua Wang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
- Department of Orthodontics, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Luwei Liu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing, China
- Department of Orthodontics, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
4
|
Cheng S, Wang KH, Zhou L, Sun ZJ, Zhang L. Tailoring Biomaterials Ameliorate Inflammatory Bone Loss. Adv Healthc Mater 2024; 13:e2304021. [PMID: 38288569 DOI: 10.1002/adhm.202304021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/08/2024] [Indexed: 05/08/2024]
Abstract
Inflammatory diseases, such as rheumatoid arthritis, periodontitis, chronic obstructive pulmonary disease, and celiac disease, disrupt the delicate balance between bone resorption and formation, leading to inflammatory bone loss. Conventional approaches to tackle this issue encompass pharmaceutical interventions and surgical procedures. Nevertheless, pharmaceutical interventions exhibit limited efficacy, while surgical treatments impose trauma and significant financial burden upon patients. Biomaterials show outstanding spatiotemporal controllability, possess a remarkable specific surface area, and demonstrate exceptional reactivity. In the present era, the advancement of emerging biomaterials has bestowed upon more efficacious solutions for combatting the detrimental consequences of inflammatory bone loss. In this review, the advances of biomaterials for ameliorating inflammatory bone loss are listed. Additionally, the advantages and disadvantages of various biomaterials-mediated strategies are summarized. Finally, the challenges and perspectives of biomaterials are analyzed. This review aims to provide new possibilities for developing more advanced biomaterials toward inflammatory bone loss.
Collapse
Affiliation(s)
- Shi Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Kong-Huai Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Lu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| |
Collapse
|
5
|
Ma H, Cai X, Hu J, Song S, Zhu Q, Zhang Y, Ma R, Shen D, Yang W, Zhou P, Zhang D, Luo Q, Hong J, Li N. Association of systemic inflammatory response index with bone mineral density, osteoporosis, and future fracture risk in elderly hypertensive patients. Postgrad Med 2024; 136:406-416. [PMID: 38753519 DOI: 10.1080/00325481.2024.2354158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVES This study sought to investigate the relationship between the systemic inflammatory response index (SIRI) and bone mineral density (BMD), osteoporosis, and future fracture risk in elderly hypertensive patients. METHODS Elderly hypertensive patients (age ≥60 years) who attended our hospital between January 2021 and December 2023 and completed BMD screening were included in the study. Analyses were performed with multivariate logistic and linear regression. RESULTS The multiple linear regression indicated that SIRI levels were significantly negatively correlated with lumbar 1 BMD (β = -0.15, 95% CI: -0.24, -0.05), lumbar 2 BMD (β = -0.15, 95% CI: -0.24, -0.05), lumbar 3 BMD (β = -1.35, 95% CI: -0.23, -0.02), lumbar 4 BMD (β = -0.11, 95% CI: -0.30, -0.10), femur neck BMD (β = -0.11, 95% CI: -0.18, -0.05) and Ward's triangle BMD (β = -0.12, 95% CI: -0.20, -0.05) among elderly hypertensive patients, after fully adjusting for confounders. Furthermore, we observed that SIRI was positively associated with future fracture risk in elderly hypertensive patients. Specifically, SIRI was associated with an increased risk of major osteoporotic fractures (β = 0.33) and hip fractures (β = 0.25). The logistic regression analysis indicated that there is an association between the SIRI level and an increased risk of osteoporosis (OR = 1.60, 95% CI = 1.37, 1.87), after fully adjusting for confounders. CONCLUSIONS Our findings indicate a potential association between SIRI and BMD, osteoporosis, and the risk of future fractures in elderly hypertensive patients. However, further studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Huimin Ma
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Xintian Cai
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Junli Hu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Shuaiwei Song
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qing Zhu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Yingying Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Rui Ma
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Di Shen
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Wenbo Yang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Pan Zhou
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Delian Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qin Luo
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Jing Hong
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Nanfang Li
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| |
Collapse
|
6
|
Liu J, Qi L, Bao S, Yan F, Chen J, Yu S, Dong C. The acute spinal cord injury microenvironment and its impact on the homing of mesenchymal stem cells. Exp Neurol 2024; 373:114682. [PMID: 38199509 DOI: 10.1016/j.expneurol.2024.114682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Spinal cord injury (SCI) is a highly debilitating condition that inflicts devastating harm on the lives of affected individuals, underscoring the urgent need for effective treatments. By activating inflammatory cells and releasing inflammatory factors, the secondary injury response creates an inflammatory microenvironment that ultimately determines whether neurons will undergo necrosis or regeneration. In recent years, mesenchymal stem cells (MSCs) have garnered increasing attention for their therapeutic potential in SCI. MSCs not only possess multipotent differentiation capabilities but also have homing abilities, making them valuable as carriers and mediators of therapeutic agents. The inflammatory microenvironment induced by SCI recruits MSCs to the site of injury through the release of various cytokines, chemokines, adhesion molecules, and enzymes. However, this mechanism has not been previously reported. Thus, a comprehensive exploration of the molecular mechanisms and cellular behaviors underlying the interplay between the inflammatory microenvironment and MSC homing is crucial. Such insights have the potential to provide a better understanding of how to harness the therapeutic potential of MSCs in treating inflammatory diseases and facilitating injury repair. This review aims to delve into the formation of the inflammatory microenvironment and how it influences the homing of MSCs.
Collapse
Affiliation(s)
- Jinyi Liu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Longju Qi
- Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Shengzhe Bao
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jiaxi Chen
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Shumin Yu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
7
|
Cai L, Lv Y, Yan Q, Guo W. Cytokines: The links between bone and the immune system. Injury 2024; 55:111203. [PMID: 38043143 DOI: 10.1016/j.injury.2023.111203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 11/05/2023] [Accepted: 11/12/2023] [Indexed: 12/05/2023]
Abstract
Osteoporosis results from an imbalance in a highly balanced physiological process called bone remodeling, in which osteoclast-mediated bone resorption and osteoblast-mediated bone formation play important roles. Osteoimmunology is a newly discovered interdisciplinary research field that focuses on the relationship between bone and the immune system. Specifically, bone and the immune system interact through cytokines, immune cells secrete cytokines, and cytokines finely regulate bone metabolism by mediating the differentiation and activity of osteoclasts and osteoblasts. Therefore, understanding the influence of cytokines on bone metabolism is conducive for the development of novel targeted drugs against immune-related bone diseases. This review summarizes the pathophysiological functions of various common cytokines in bone and discusses the potential clinical value of multiple cytokines in immune-mediated bone diseases.
Collapse
Affiliation(s)
- Liping Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China; Department of Endocrinology, Rheumatology and Immunology, Anyang People's Hospital, Anyang, Henan 455000, China
| | - You Lv
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Qihui Yan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
8
|
Sheng N, Xing F, Wang J, Zhang QY, Nie R, Li-Ling J, Duan X, Xie HQ. Recent progress in bone-repair strategies in diabetic conditions. Mater Today Bio 2023; 23:100835. [PMID: 37928253 PMCID: PMC10623372 DOI: 10.1016/j.mtbio.2023.100835] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/02/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023] Open
Abstract
Bone regeneration following trauma, tumor resection, infection, or congenital disease is challenging. Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia. It can result in complications affecting multiple systems including the musculoskeletal system. The increased number of diabetes-related fractures poses a great challenge to clinical specialties, particularly orthopedics and dentistry. Various pathological factors underlying DM may directly impair the process of bone regeneration, leading to delayed or even non-union of fractures. This review summarizes the mechanisms by which DM hampers bone regeneration, including immune abnormalities, inflammation, reactive oxygen species (ROS) accumulation, vascular system damage, insulin/insulin-like growth factor (IGF) deficiency, hyperglycemia, and the production of advanced glycation end products (AGEs). Based on published data, it also summarizes bone repair strategies in diabetic conditions, which include immune regulation, inhibition of inflammation, reduction of oxidative stress, promotion of angiogenesis, restoration of stem cell mobilization, and promotion of osteogenic differentiation, in addition to the challenges and future prospects of such approaches.
Collapse
Affiliation(s)
- Ning Sheng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Fei Xing
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jie Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Duan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| |
Collapse
|
9
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
10
|
Choa R, Panaroni C, Bhatia R, Raje N. It is worth the weight: obesity and the transition from monoclonal gammopathy of undetermined significance to multiple myeloma. Blood Adv 2023; 7:5510-5523. [PMID: 37493975 PMCID: PMC10515310 DOI: 10.1182/bloodadvances.2023010822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
The overweight/obesity epidemic is a serious public health concern that affects >40% of adults globally and increases the risk of numerous chronic diseases, such as type 2 diabetes, heart disease, and various cancers. Multiple myeloma (MM) is a lymphohematopoietic cancer caused by the uncontrolled clonal expansion of plasma cells. Recent studies have shown that obesity is a risk factor not only for MM but also monoclonal gammopathy of undetermined significance (MGUS), a precursor disease state of MM. Furthermore, obesity may promote the transition from MGUS to MM. Thus, in this review, we summarize the epidemiological evidence regarding the role of obesity in MM and MGUS, discuss the biologic mechanisms that drive these disease processes, and detail the obesity-targeted pharmacologic and lifestyle interventions that may reduce the risk of progression from MGUS to MM.
Collapse
Affiliation(s)
- Ruth Choa
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Cristina Panaroni
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Roma Bhatia
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Noopur Raje
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
11
|
Xu J, Yu L, Liu F, Wan L, Deng Z. The effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis: a review. Front Immunol 2023; 14:1222129. [PMID: 37475866 PMCID: PMC10355373 DOI: 10.3389/fimmu.2023.1222129] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/12/2023] [Indexed: 07/22/2023] Open
Abstract
The complicated connections and cross talk between the skeletal system and the immune system are attracting more attention, which is developing into the field of Osteoimmunology. In this field, cytokines that are among osteoblasts and osteoclasts play a critical role in bone remodeling, which is a pathological process in the pathogenesis and development of osteoporosis. Those cytokines include the tumor necrosis factor (TNF) family, the interleukin (IL) family, interferon (IFN), chemokines, and so on, most of which influence the bone microenvironment, osteoblasts, and osteoclasts. This review summarizes the effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis, aiming to providing the latest reference to the role of immunology in osteoporosis.
Collapse
Affiliation(s)
- Jie Xu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linxin Yu
- Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Longbiao Wan
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Deng
- Hubei Provincial Hospital of Traditional Chinese Medicine (TCM), Wuhan, China
| |
Collapse
|
12
|
Li S, Zhang L, Liu C, Kim J, Su K, Chen T, Zhao L, Lu X, Zhang H, Cui Y, Cui X, Yuan F, Pan H. Spontaneous immunomodulation and regulation of angiogenesis and osteogenesis by Sr/Cu-borosilicate glass (BSG) bone cement to repair critical bone defects. Bioact Mater 2023; 23:101-117. [DOI: 10.1016/j.bioactmat.2022.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
|
13
|
Abstract
Bone marrow contains resident cellular components that are not only involved in bone maintenance but also regulate hematopoiesis and immune responses. The immune system and bone interact with each other, coined osteoimmunology. Hashimoto's thyroiditis (HT) is one of the most common chronic autoimmune diseases which is accompanied by lymphocytic infiltration. It shows elevating thyroid autoantibody levels at an early stage and progresses to thyroid dysfunction ultimately. Different effects exert on bone metabolism during different phases of HT. In this review, we summarized the mechanisms of the long-term effects of HT on bone and the relationship between thyroid autoimmunity and osteoimmunology. For patients with HT, the bone is affected not only by thyroid function and the value of TSH, but also by the setting of the autoimmune background. The autoimmune background implies a breakdown of the mechanisms that control self-reactive system, featuring abnormal immune activation and presence of autoantibodies. The etiology of thyroid autoimmunity and osteoimmunology is complex and involves a number of immune cells, cytokines and chemokines, which regulate the pathogenesis of HT and osteoporosis at the same time, and have potential to affect each other. In addition, vitamin D works as a potent immunomodulator to influence both thyroid immunity and osteoimmunology. We conclude that HT affects bone metabolism at least through endocrine and immune pathways.
Collapse
Affiliation(s)
- Jialu Wu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, 37 Guoxue Lane, 610041, Chengdu, P.R. China
| | - Hui Huang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, 37 Guoxue Lane, 610041, Chengdu, P.R. China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, 37 Guoxue Lane, 610041, Chengdu, P.R. China.
| |
Collapse
|
14
|
Jiang Z, Jin L, Jiang C, Yan Z, Cao Y. IL-1β contributes to the secretion of sclerostin by osteocytes and targeting sclerostin promotes spinal fusion at early stages. J Orthop Surg Res 2023; 18:162. [PMID: 36864451 PMCID: PMC9983224 DOI: 10.1186/s13018-023-03657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 02/28/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Despite extensive research, there is still a need for safe and effective agents to promote spinal fusion. Interleukin (IL)-1β is an important factor which influences the bone repair and remodelling. The purpose of our study was to determine the effect of IL-1β on sclerostin in osteocytes and to explore whether inhibiting the secretion of sclerostin from osteocytes can promote spinal fusion at early stages. METHODS Small-interfering RNA was used to suppress the secretion of sclerostin in Ocy454 cells. MC3T3-E1 cells were cocultured with Ocy454 cells. Osteogenic differentiation and mineralisation of MC3T3-E1 cells were evaluated in vitro. SOST knock-out rat generated using the CRISPR-Cas9 system and rat spinal fusion model was used in vivo. The degree of spinal fusion was assessed by manual palpation, radiographic analysis and histological analysis at 2 and 4 weeks. RESULTS We found that IL-1β level had a positive association with sclerostin level in vivo. IL-1β promoted the expression and secretion of sclerostin in Ocy454 cells in vitro. Inhibition of IL-1β-induced secretion of sclerostin from Ocy454 cells could promote the osteogenic differentiation and mineralisation of cocultured MC3T3-E1 cells in vitro. The extent of spinal graft fusion was greater in SOST-knockout rats than in wild-type rats at 2 and 4 weeks. CONCLUSIONS The results demonstrate that IL-1β contributes to a rise in the level of sclerostin at early stages of bone healing. Suppressing sclerostin may be an important therapeutic target capable of promoting spinal fusion at early stages.
Collapse
Affiliation(s)
- Zengxin Jiang
- Department of Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China.,Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, NO. 180 Feng Lin Road, Xuhui District, Shanghai, 200032, China
| | - Lixia Jin
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, NO. 180 Feng Lin Road, Xuhui District, Shanghai, 200032, China
| | - Chang Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, NO. 180 Feng Lin Road, Xuhui District, Shanghai, 200032, China
| | - Zuoqin Yan
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, NO. 180 Feng Lin Road, Xuhui District, Shanghai, 200032, China.
| | - Yuanwu Cao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, NO. 180 Feng Lin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
15
|
Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells 2023; 12:cells12040521. [PMID: 36831188 PMCID: PMC9954309 DOI: 10.3390/cells12040521] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.
Collapse
|
16
|
Gao Y, Min Q, Li X, Liu L, Lv Y, Xu W, Liu X, Wang H. Immune System Acts on Orthodontic Tooth Movement: Cellular and Molecular Mechanisms. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9668610. [PMID: 36330460 PMCID: PMC9626206 DOI: 10.1155/2022/9668610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/05/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022]
Abstract
Orthodontic tooth movement (OTM) is a tissue remodeling process based on orthodontic force loading. Compressed periodontal tissues have a complicated aseptic inflammatory cascade, which are considered the initial factor of alveolar bone remodeling. Since skeletal and immune systems shared a wide variety of molecules, osteoimmunology has been generally accepted as an interdisciplinary field to investigate their interactions. Unsurprisingly, OTM is considered a good mirror of osteoimmunology since it involves immune reaction and bone remolding. In fact, besides bone remodeling, OTM involves cementum resorption, soft tissue remodeling, orthodontic pain, and relapse, all correlated with immune cells and/or immunologically active substance. The aim of this paper is to review the interaction of immune system with orthodontic tooth movement, which helps gain insights into mechanisms of OTM and search novel method to short treatment period and control complications.
Collapse
Affiliation(s)
- Yajun Gao
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Qingqing Min
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Xingjia Li
- Department of Prosthodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Linxiang Liu
- Department of Implantology, Wuxi Stomatology Hospital, Wuxi, China
| | - Yangyang Lv
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Wenjie Xu
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | | | - Hua Wang
- Wuhu Stomatology Hospital, Wuhu, China
| |
Collapse
|
17
|
Cao JF, Hu X, Xiong L, Wu M, Yang X, Wang C, Chen S, Xu H, Chen H, Ma X, Mi Y, Zhang X. Interference of Interleukin-1 β Mediated by Lentivirus Promotes Functional Recovery of Spinal Cord Contusion Injury in Rats via the PI3K/AKT1 Signaling Pathway. Mediators Inflamm 2022; 2022:6285099. [PMID: 39262872 PMCID: PMC11390212 DOI: 10.1155/2022/6285099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/30/2022] [Indexed: 09/13/2024] Open
Abstract
Purpose Inflammation and apoptosis after spinal cord contusion (SCC) are important causes of irreversible spinal cord injury. Interleukin-1β (IL-1β) is a key inflammatory factor that promotes the aggravation of spinal cord contusion. However, the specific role and regulatory mechanism of IL-1β in spinal cord contusion is still unclear. Therefore, this study applied bioinformatics to analyze and mine potential gene targets interlinked with IL-1β, animal experiments and lentiviral interference technology were used to explore whether IL-1β affected the recovery of motor function in spinal cord contusion by interfering with PI3K/AKT1 signaling pathway. Method This study used bioinformatics to screen and analyze gene targets related to IL-1β. The rat SCC animal model was established by the Allen method, and the Basso Beattie Bresnahan (BBB) score was used to evaluate the motor function of the spinal cord-injured rats. Immunohistochemistry and immunofluorescence were used to localize the expression of IL-1β and AKT1 proteins in spinal cord tissue. Quantitative polymerase chain reaction and Western blot were used to detect the gene and protein expressions of IL-1β, PI3K, and AKT1. RNAi technology was used to construct lentivirus to inhibit the expression of IL-1β, lentiviral interference with IL-1β was used to investigate the effect of IL-1β and AKT1 on the function of spinal cord contusion and the relationship among IL-1β, AKT1, and downstream signaling pathways. Results Bioinformatics analysis suggested a close relationship between IL-1β and AKT1. Animal experiments have confirmed that IL-1β is closely related to the functional recovery of spinal cord contusion. Firstly, from the phenomenological level, the BBB score decreased after SCC, IL-1β and AKT1 were located in the cytoplasm of neurons in the anterior horn of the spinal cord, and the expression levels of IL-1β gene and protein in the experimental group were higher than those in the sham operation group. At the same time, the expression of AKT1 gene decreased, the results suggested that the increase of IL-1β affected the functional recovery of spinal cord contusion. Secondly, from the functional level, after inhibiting the expression of IL-1β with a lentivirus-mediated method, the BBB score was significantly increased, and the motor function of the spinal cord was improved. Thirdly, from the mechanistic level, bioinformatics analysis revealed the relationship between IL-1β and AKT1. In addition, the experiment further verified that in the PI3K/AKT1 signaling pathway, inhibition of IL-1β expression upregulated AKT1 gene expression, but PI3K expression was unchanged. Conclusion Inhibition of IL-1β promotes recovery of motor function after spinal cord injury in rats through upregulation of AKT1 expression in the PI3K/AKT1 signaling pathway. Bioinformatics analysis suggested that IL-1β may affect apoptosis and regeneration by inhibiting the expression of AKT1 in the PI3K/AKT1 signaling pathway to regulate the downstream FOXO, mTOR, and GSK3 signaling pathways; thereby hindering the recovery of motor function in rats after spinal cord contusion. It provided a new perspective for clinical treatment of spinal cord contusion in the future.
Collapse
Affiliation(s)
- Jun-Feng Cao
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xi Hu
- Taikang Tongji Wuhan Hospital, Wuhan, China
| | - Li Xiong
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Mei Wu
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Xingyu Yang
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Chaochao Wang
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Shengyan Chen
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Hengxiang Xu
- Clinical Medical College of Chengdu Medical College, Chengdu, China
| | - Huanyu Chen
- Basic Medical College of Chengdu Medical College, Chengdu, China
| | - Xuntai Ma
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yongjie Mi
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- National Demonstration Center for Experimental Clinical Medicine Education of Chengdu Medical College, Chengdu, China
| | - Xiao Zhang
- Clinical Medical College of Chengdu Medical College, Chengdu, China
- Basic Medical College of Chengdu Medical College, Chengdu, China
- National Demonstration Center for Experimental Clinical Medicine Education of Chengdu Medical College, Chengdu, China
| |
Collapse
|
18
|
Xu W, Jiang Y, Wang N, Bai H, Xu S, Xia T, Xin H. Traditional Chinese Medicine as a Promising Strategy for the Treatment of Alzheimer's Disease Complicated With Osteoporosis. Front Pharmacol 2022; 13:842101. [PMID: 35721142 PMCID: PMC9198449 DOI: 10.3389/fphar.2022.842101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) and osteoporosis (OP) are progressive degenerative diseases caused by multiple factors, placing a huge burden on the world. Much evidence indicates that OP is a common complication in AD patients. In addition, there is also evidence to show that patients with OP have a higher risk of AD than those without OP. This suggests that the association between the two diseases may be due to a pathophysiological link rather than one disease causing the other. Several in vitro and in vivo studies have also proved their common pathogenesis. Based on the theory of traditional Chinese medicine, some classic and specific natural Chinese medicines are widely used to effectively treat AD and OP. Current evidence also shows that these treatments can ameliorate both brain damage and bone metabolism disorder and further alleviate AD complicated with OP. These valuable therapies might provide effective and safe alternatives to major pharmacological strategies.
Collapse
Affiliation(s)
- Weifan Xu
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China.,Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yiping Jiang
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Nani Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Huanhuan Bai
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Shengyan Xu
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Tianshuang Xia
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| | - Hailiang Xin
- Department of Pharmacognosy, School of Pharmacy, Naval Medical University, Shanghai, China
| |
Collapse
|
19
|
Tan WH, Winkler C. A non-disruptive and efficient knock-in approach allows fate tracing of resident osteoblast progenitors during repair of vertebral lesions in medaka. Development 2022; 149:275483. [DOI: 10.1242/dev.200238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 05/11/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
During bone development and repair, osteoblasts are recruited to bone deposition sites. To identify the origin of recruited osteoblasts, cell lineage tracing using Cre/loxP recombination is commonly used. However, a confounding factor is the use of transgenic Cre drivers that do not accurately recapitulate endogenous gene expression or the use of knock-in Cre drivers that alter endogenous protein activity or levels. Here, we describe a CRISPR/Cas9 homology-directed repair knock-in approach that allows efficient generation of Cre drivers controlled by the endogenous gene promoter. In addition, a self-cleaving peptide preserves the reading frame of the endogenous protein. Using this approach, we generated col10a1p2a-CreERT2 knock-in medaka and show that tamoxifen-inducible CreERT2 efficiently recombined loxP sites in col10a1 cells. Similar knock-in efficiencies were obtained when two unrelated loci (osr1 and col2a1a) were targeted. Using live imaging, we traced the fate of col10a1 osteoblast progenitors during bone lesion repair in the medaka vertebral column. We show that col10a1 cells at neural arches represent a mobilizable cellular source for bone repair. Together, our study describes a previously unreported strategy for precise cell lineage tracing via efficient and non-disruptive knock-in of Cre.
Collapse
Affiliation(s)
- Wen Hui Tan
- National University of Singapore Department of Biological Sciences and Centre for Bioimaging Sciences , , Singapore 117543 , Singapore
| | - Christoph Winkler
- National University of Singapore Department of Biological Sciences and Centre for Bioimaging Sciences , , Singapore 117543 , Singapore
| |
Collapse
|
20
|
Bordukalo-Nikšić T, Kufner V, Vukičević S. The Role Of BMPs in the Regulation of Osteoclasts Resorption and Bone Remodeling: From Experimental Models to Clinical Applications. Front Immunol 2022; 13:869422. [PMID: 35558080 PMCID: PMC9086899 DOI: 10.3389/fimmu.2022.869422] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
In response to mechanical forces and the aging process, bone in the adult skeleton is continuously remodeled by a process in which old and damaged bone is removed by bone-resorbing osteoclasts and subsequently is replaced by new bone by bone-forming cells, osteoblasts. During this essential process of bone remodeling, osteoclastic resorption is tightly coupled to osteoblastic bone formation. Bone-resorbing cells, multinuclear giant osteoclasts, derive from the monocyte/macrophage hematopoietic lineage and their differentiation is driven by distinct signaling molecules and transcription factors. Critical factors for this process are Macrophage Colony Stimulating Factor (M-CSF) and Receptor Activator Nuclear Factor-κB Ligand (RANKL). Besides their resorption activity, osteoclasts secrete coupling factors which promote recruitment of osteoblast precursors to the bone surface, regulating thus the whole process of bone remodeling. Bone morphogenetic proteins (BMPs), a family of multi-functional growth factors involved in numerous molecular and signaling pathways, have significant role in osteoblast-osteoclast communication and significantly impact bone remodeling. It is well known that BMPs help to maintain healthy bone by stimulating osteoblast mineralization, differentiation and survival. Recently, increasing evidence indicates that BMPs not only help in the anabolic part of bone remodeling process but also significantly influence bone catabolism. The deletion of the BMP receptor type 1A (BMPRIA) in osteoclasts increased osteoblastic bone formation, suggesting that BMPR1A signaling in osteoclasts regulates coupling to osteoblasts by reducing bone-formation activity during bone remodeling. The dual effect of BMPs on bone mineralization and resorption highlights the essential role of BMP signaling in bone homeostasis and they also appear to be involved in pathological processes in inflammatory disorders affecting bones and joints. Certain BMPs (BMP2 and -7) were approved for clinical use; however, increased bone resorption rather than formation were observed in clinical applications, suggesting the role BMPs have in osteoclast activation and subsequent osteolysis. Here, we summarize the current knowledge of BMP signaling in osteoclasts, its role in osteoclast resorption, bone remodeling, and osteoblast–osteoclast coupling. Furthermore, discussion of clinical application of recombinant BMP therapy is based on recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Tatjana Bordukalo-Nikšić
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vera Kufner
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Slobodan Vukičević
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
21
|
Ou P, Zhang T, Wang J, Li C, Shao C, Ruan J. Microstructure, mechanical properties and osseointegration ability of Ta-20Zr alloy used as dental implant material. Biomed Mater 2022; 17:045003. [PMID: 35477054 DOI: 10.1088/1748-605x/ac6b05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/27/2022] [Indexed: 11/12/2022]
Abstract
The aim of this study was to evaluate the application prospect of a tantalum (Ta) and zirconium (Zr) alloy as a dental implant material. The Ta-20Zr (wt.%) alloy was prepared by powder metallurgy, and its microstructure and mechanical properties were analyzed by standard techniques. The effect of Ta-20Zr alloy on inflammation, bone remodeling and osseointegration was analyzed in rat and rabbit models by biochemical, histological and imaging tests. The Ta-20Zr alloy showed excellent mechanical compatibility with the bone tissue on account of similar elastic modulus (49.2 GPa), thereby avoiding the 'stress shielding effect'. Furthermore, Ta-20Zr alloy enhanced the inflammatory response by promoting secretion of interleukin-6 (IL-6) and IL-10, and facilitated the balance between the M1/M2 macrophage phenotypes. Finally, Ta-20Zr also showed excellent osseointegration and osteogenic ability without any systemic side effects, making it an ideal dental implant material.
Collapse
Affiliation(s)
- Pinghua Ou
- Department of Stomatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan, 410083, People's Republic of China
| | - Taomei Zhang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan, 410083, People's Republic of China
| | - Jianying Wang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan, 410083, People's Republic of China
| | - Cui Li
- Department of Stomatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Chunsheng Shao
- Department of Stomatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Jianming Ruan
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, Hunan, 410083, People's Republic of China
| |
Collapse
|
22
|
Srivastava RK, Sapra L. The Rising Era of “Immunoporosis”: Role of Immune System in the Pathophysiology of Osteoporosis. J Inflamm Res 2022; 15:1667-1698. [PMID: 35282271 PMCID: PMC8906861 DOI: 10.2147/jir.s351918] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/10/2022] [Indexed: 12/21/2022] Open
Abstract
Discoveries in the last few years have emphasized the existence of an enormous breadth of communication between bone and the immune system in maintaining skeletal homeostasis. Originally, the discovery of various factors was assigned to the immune system viz. interleukin (IL)-6, IL-10, IL-17, tumor necrosis factor (TNF)-α, receptor activator of nuclear factor kappa B ligand (RANKL), nuclear factor of activated T cells (NFATc1), etc., but now these factors have also been shown to have a significant impact on osteoblasts (OBs) and osteoclasts (OCs) biology. These discoveries led to an alteration in the approach for the treatment of several bone pathologies including osteoporosis. Osteoporosis is an inflammatory bone anomaly affecting more than 500 million people globally. In 2018, to highlight the importance of the immune system in the pathophysiology of osteoporosis, our group coined the term “immunoporosis”. In the present review, we exhaustively revisit the characteristics, mechanism of action, and function of both innate and adaptive immune cells with the goal of understanding the potential of immune cells in osteoporosis. We also highlight the Immunoporotic role of gut microbiota (GM) for the treatment and management of osteoporosis. Importantly, we further discuss whether an immune cell-based strategy to treat and manage osteoporosis is feasible and relevant in clinical settings.
Collapse
Affiliation(s)
- Rupesh K Srivastava
- Immunoporosis Lab, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
- Correspondence: Rupesh K Srivastava, Tel +91 11-26593548, Email ;
| | - Leena Sapra
- Immunoporosis Lab, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| |
Collapse
|
23
|
Myeloproliferative Disorders and its Effect on Bone Homeostasis: The Role of Megakaryocytes. Blood 2021; 139:3127-3137. [PMID: 34428274 DOI: 10.1182/blood.2021011480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
Myeloproliferative Neoplasms (MPNs) are a heterogeneous group of chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells, of which Polycythemia Vera (PV), Essential Thrombocythemia (ET), and Primary Myelofibrosis (PMF) have been extensively reviewed in context of clonal expansion, fibrosis and other phenotypes. Here, we review current knowledge on the influence of different forms of MPN on bone health. Studies implicated various degrees of effect of different forms of MPN on bone density, and on osteoblast proliferation and differentiation, using murine models and human data. The majority of studies show that bone volume is generally increased in PMF patients, whereas it is slightly decreased or not altered in ET and PV patients, although possible differences between male and female phenotypes were not fully explored in most MPN forms. Osteosclerosis seen in PMF patients is a serious complication that can lead to bone marrow failure, and the loss of bone reported in some ET and PV patients can lead to osteoporotic fractures. Some MPN forms are associated with increased number of megakaryocytes (MKs), and several of the MK-associated factors in MPN are known to affect bone development. Here, we review known mechanisms involved in these processes, with focus on the role of MKs and secreted factors. Understanding MPN-associated changes in bone health could improve early intervention and treatment of this side effect of the pathology.
Collapse
|
24
|
Li H, He D, Xiao X, Yu G, Hu G, Zhang W, Wen X, Lin Y, Li X, Lin H, Diao Y, Tang Y. Nitrogen-Doped Multiwalled Carbon Nanotubes Enhance Bone Remodeling through Immunomodulatory Functions. ACS APPLIED MATERIALS & INTERFACES 2021; 13:25290-25305. [PMID: 33908252 DOI: 10.1021/acsami.1c05437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
It has been reported that multiwalled carbon nanotubes (MWCNTs) can reportedly positively affect growth and differentiation of bone-related cells and therefore offer great potential in biomedical applications. To overcome negative immune responses that limit their application, specific doping and functionalization can improve their biocompatibility. Here, we demonstrated that nitrogen-doped carboxylate-functionalized MWCNTs (N-MWCNTs) enhance bone remodeling both in vitro and in vivo with excellent biocompatibility, via stimulation of both bone resorption and formation. We revealed that 0.2 μg/mL N-MWCNTs not only increase the transcription of osteoblastogenic and osteoclastogenic genes but also up-regulate the activities of both TRAP and AKP in the differentiation of bone marrow stromal cells (BMSCs). Additionally, intramuscular administration of N-MWCNTs at a dosage of 1.0 mg/kg body weight enhances bone mineral density and bone mass content in mice, as well as induces potentiated degree of TRAP- and ARS-positive staining in the femur. The positive regulation of N-MWCNTs on bone remodeling is initiated by macrophage phagocytosis, which induces altered production of inflammatory cytokines by immune response pathways, and consequently up-regulates IL1α, IL10, and IL16. These cytokines collectively regulate the central osteoclastogenic transcription factor NFATc1 and osteoblastogenic BMP signaling, the suppression of which confirmed that these factors respectively participate in N-MWCNT-mediated regulation of osteoclastic and osteoblastic bone marrow stem cell activities. These results suggest that N-MWCNTs can be readily generalized for use as biomaterials in bone tissue engineering for metabolic bone disorders.
Collapse
Affiliation(s)
- Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Dalin He
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Xue Xiao
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Guanliu Yu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Geng Hu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Wenqian Zhang
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Xin Wen
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Yun Lin
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Xianyao Li
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Hai Lin
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Youxiang Diao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| | - Yi Tang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an 271018, China
| |
Collapse
|
25
|
Ni J, Wu Z. Inflammation Spreading: Negative Spiral Linking Systemic Inflammatory Disorders and Alzheimer's Disease. Front Cell Neurosci 2021; 15:638686. [PMID: 33716675 PMCID: PMC7947253 DOI: 10.3389/fncel.2021.638686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022] Open
Abstract
As a physiological response to injury in the internal body organs, inflammation is responsible for removing dangerous stimuli and initiating healing. However, persistent and exaggerative chronic inflammation causes undesirable negative effects in the organs. Inflammation occurring in the brain and spinal cord is known as neuroinflammation, with microglia acting as the central cellular player. There is increasing evidence suggesting that chronic neuroinflammation is the most relevant pathological feature of Alzheimer’s disease (AD), regulating other pathological features, such as the accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau. Systemic inflammatory signals caused by systemic disorders are known to strongly influence neuroinflammation as a consequence of microglial activation, inflammatory mediator production, and the recruitment of peripheral immune cells to the brain, resulting in neuronal dysfunction. However, the neuroinflammation-accelerated neuronal dysfunction in AD also influences the functions of peripheral organs. In the present review, we highlight the link between systemic inflammatory disorders and AD, with inflammation serving as the common explosion. We discuss the molecular mechanisms that govern the crosstalk between systemic inflammation and neuroinflammation. In our view, inflammation spreading indicates a negative spiral between systemic diseases and AD. Therefore, “dampening inflammation” through the inhibition of cathepsin (Cat)B or CatS may be a novel therapeutic approach for delaying the onset of and enacting early intervention for AD.
Collapse
Affiliation(s)
- Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.,OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Venugopal M, Nambiar J, Nair BG. Anacardic acid-mediated regulation of osteoblast differentiation involves mitigation of inflammasome activation pathways. Mol Cell Biochem 2020; 476:819-829. [PMID: 33090336 DOI: 10.1007/s11010-020-03947-9] [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: 07/19/2020] [Accepted: 10/10/2020] [Indexed: 10/23/2022]
Abstract
Disruption of the finely tuned osteoblast-osteoclast balance is the underlying basis of several inflammatory bone diseases, such as osteomyelitis, osteoporosis, and septic arthritis. Prolonged and unrestrained exposure to inflammatory environment results in reduction of bone mineral density by downregulating osteoblast differentiation. Earlier studies from our laboratory have identified that Anacardic acid (AA), a constituent of Cashew nut shell liquid that is used widely in traditional medicine, has potential inhibitory effect on gelatinases (MMP2 and MMP9) which are over-expressed in numerous inflammatory conditions (Omanakuttan et al. in Mol Pharmacol, 2012 and Nambiar et al. in Exp Cell Res, 2016). The study demonstrated for the first time that AA promotes osteoblast differentiation in lipopolysaccharide-treated osteosarcoma cells (MG63) by upregulating specific markers, like osteocalcin, receptor activator of NF-κB ligand, and alkaline phosphatase. Furthermore, expression of the negative regulators, such as nuclear factor-κB, matrix metalloproteinases (MMPs), namely MMP13, and MMP1, along with several inflammatory markers, such as Interleukin-1β and Nod-like receptor protein 3 were downregulated by AA. Taken together, AA expounds as a novel template for development of potential pharmacological therapeutics for inflammatory bone diseases.
Collapse
Affiliation(s)
- Meera Venugopal
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, India, 690525
| | - Jyotsna Nambiar
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, India, 690525
| | - Bipin G Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O, Kollam, Kerala, India, 690525.
| |
Collapse
|
27
|
The Effects of Titanium Surfaces Modified with an Antimicrobial Peptide GL13K by Silanization on Polarization, Anti-Inflammatory, and Proinflammatory Properties of Macrophages. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2327034. [PMID: 32775410 PMCID: PMC7396038 DOI: 10.1155/2020/2327034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/04/2020] [Accepted: 06/25/2020] [Indexed: 11/23/2022]
Abstract
The polarization of macrophages and its anti-inflammatory and proinflammatory properties play a significant role in host response after implant placement to determine the outcome of osseointegration and long-term survival. In the previous study, we immobilized an antimicrobial peptide, GL13K, onto titanium surfaces to provide immune regulation property. In the herein presented study, we aimed at investigating whether GL13K immobilized titanium surface could improve osteogenesis and reduce the inflammatory reaction around the biomaterials by altering macrophage response. We evaluated the cell proliferation of the different phenotypes of macrophages seeded in GL13K-coated titanium surface, which indicated an inhibition of M1 macrophages and a good cytocompatibility to M2 macrophages. Then, we measured the inflammatory and anti-inflammatory activity of the M1 and M2 macrophages seeded on the GL13K-coated titanium surfaces. The results of the enzyme-linked immunosorbent assay and quantitative reverse transcription-polymerase chain reaction showed that the group with the GL13K modified surface had a downregulation in the expression level of the tumor necrosis factor-α and interleukin-1β in M1 macrophages and an upregulation of IL-10 and transforming growth factor-β3 (TGF-β3) levels in M2 macrophages. This study demonstrated that the GL13K modified titanium surfaces can regulate macrophages' polarization and the expression of inflammatory and anti-inflammatory effects, reducing the effects of the inflammatory process, which may promote the process of bone regeneration and osseointegration.
Collapse
|
28
|
Yalçınozan M, Türker M, Çırpar M. Effects of a novel biodegredable implant system on a rat tibia fracture model. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2020; 54:453-460. [PMID: 32812878 DOI: 10.5152/j.aott.2020.18331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aimed to determine the effects of a novel biodegradable implant releasing platelet-derived growth factor (PDGF) at the fracture site on fracture healing in a rat tibia fracture model. METHODS In this study, 35 male Sprague-Dawley rats weighing between 300 and 350 g were used. The rats were divided into four groups: Group A (control group without any treatment, n=10), Group B (spacer without PDGF Group, n=10), Group C (spacer with PDGF group, n=10), and Group D (healthy rat Group, n=5). Standardized fractures were created in the right tibias of rats, and then biodegradable implants made of poly-β-hydroxybutyrate-co-3-hydroxy valerate were implanted at the fracture sites in Groups B and C. In Group C, implants were loaded with 600 ng of PDGF. Animals were sacrificed 30 days after the operation, and fracture healing in each group was assessed radiologically based on the Goldberg score. Furthermore, the anteroposterior (AP) and mediolateral (ML) callus diameters were measured macroscopically, and fracture sites were mechanically tested. RESULTS In the radiological assessment, Group C showed higher fracture healing rate than Groups A and B (p=0.001), whereas no significant difference was found between group C and Group D (p>0.05). In the macroscopic assessment, while Group C exhibited the thickest AP callus diameter (p=0.02), no significant differences in ML callus diameters existed among the groups (p>0.05). Mechanical testing revealed that Group C had higher torsional strength (p=0.001) and stiffness than Groups A and B (p=0.001) while there was no significant difference between Groups C and D (p>0.05). CONCLUSION Biodegradable implant releasing PDGF may have positive effects on fracture healing.
Collapse
Affiliation(s)
- Mehmet Yalçınozan
- Department of Orthopaedics and Traumatology, Near East University Hospital, Mersin, Turkey
| | - Mehmet Türker
- Department of Orthopaedics and Traumatology, Sakarya University School of Medicine, Sakarya, Turkey
| | - Meriç Çırpar
- Department of Orthopaedics and Traumatology, Kırıkkale University School of Medicine, Kırıkkale, Turkey
| |
Collapse
|
29
|
Kawabata T, Tokuda H, Kuroyanagi G, Fujita K, Sakai G, Kim W, Matsushima-Nishiwaki R, Iida H, Yata KI, Wang S, Mizoguchi A, Otsuka T, Kozawa O. Incretin accelerates platelet-derived growth factor-BB-induced osteoblast migration via protein kinase A: The upregulation of p38 MAP kinase. Sci Rep 2020; 10:2341. [PMID: 32047216 PMCID: PMC7012849 DOI: 10.1038/s41598-020-59392-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 01/27/2020] [Indexed: 11/18/2022] Open
Abstract
Incretins, including glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), secreted from enteroendocrine cells after food ingestion, are currently recognized to regulate glucose metabolism through insulin secretion. We previously demonstrated that platelet-derived growth factor-BB (PDGF-BB) induces the migration of osteoblast-like MC3T3-E1 cells through mitogen-activated protein (MAP) kinases, including p38 MAP kinase. In the present study, we investigated whether or not incretins affect the osteoblast migration. The PDGF-BB-induced cell migration was significantly reinforced by GLP-1, GIP or cAMP analogues in MC3T3-E1 cells and normal human osteoblasts. The upregulated migration by GLP-1 or cAMP analogues was suppressed by H89, an inhibitor of protein kinase A. The amplification by GLP-1 of migration induced by PDGF-BB was almost completely reduced by SB203580, a p38 MAP kinase inhibitor in MC3T3-E1 cells and normal human osteoblasts. In addition, GIP markedly strengthened the PDGF-BB-induced phosphorylation of p38 MAP kinase. Exendin-4, a GLP-1 analogue, induced Rho A expression and its translocation from cytoplasm to plasma membranes in osteoblasts at the epiphyseal lines of developing mouse femurs in vivo. These results strongly suggest that incretins accelerates the PDGF-BB-induced migration of osteoblasts via protein kinase A, and the up-regulation of p38 MAP kinase is involved in this acceleration. Our findings may highlight the novel potential of incretins to bone physiology and therapeutic strategy against bone repair.
Collapse
Affiliation(s)
- Tetsu Kawabata
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan.,Department of Orthopedic Surgery, Toyokawa City Hospital, Toyokawa, 442-8561, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Clinical Laboratory/Medical Genome Center Biobank, National Center for Geriatrics and Gerontology, Obu, 474-8511, Japan
| | - Gen Kuroyanagi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Rehabilitation Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Kazuhiko Fujita
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Go Sakai
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Woo Kim
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | | | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Ken-Ichiro Yata
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, 514-8507, Japan
| | - Shujie Wang
- Deaprtment of Neural Regeneration and Cell Communication, Graduate School of Medicine, Mie University, Tsu, 514-8507, Japan
| | - Akira Mizoguchi
- Deaprtment of Neural Regeneration and Cell Communication, Graduate School of Medicine, Mie University, Tsu, 514-8507, Japan
| | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.
| |
Collapse
|
30
|
Abstract
Cytokines and hematopoietic growth factors have traditionally been thought of as regulators of the development and function of immune and blood cells. However, an ever-expanding number of these factors have been discovered to have major effects on bone cells and the development of the skeleton in health and disease (Table 1). In addition, several cytokines have been directly linked to the development of osteoporosis in both animal models and in patients. In order to understand the mechanisms regulating bone cells and how this may be dysregulated in disease states, it is necessary to appreciate the diverse effects that cytokines and inflammation have on osteoblasts, osteoclasts, and bone mass. This chapter provides a broad overview of this topic with extensive references so that, if desired, readers can access specific references to delve into individual topics in greater detail.
Collapse
Affiliation(s)
- Joseph Lorenzo
- Departments of Medicine and Orthopaedic Surgery, UConn Health, Farmington, CT, USA.
| |
Collapse
|
31
|
Inflammatory markers and bone mass in children with overweight/obesity: the role of muscular fitness. Pediatr Res 2020; 87:42-47. [PMID: 31493774 DOI: 10.1038/s41390-019-0572-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/02/2019] [Accepted: 08/16/2019] [Indexed: 11/09/2022]
Abstract
OBJECTIVES To examine which inflammatory markers are associated with bone mass and whether this association varies according to muscular fitness in children with overweight/obesity. METHODS Plasma interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), epidermal growth factor, vascular endothelial growth factor A (VEGF), and C-reactive protein were analyzed in 55 children aged 8-11 years. A muscular fitness score was computed. Bone mineral content (BMC) of the total body-less head (TBLH) and lumbar spine (LS) were assessed using dual-energy x-ray absorptiometry. RESULTS IL-6 (β = -0.136) and VEGF (β = -0.099) were associated with TBLH BMC, while TNF-α (β = -0.345) and IL-1β (β = 0.212) were associated with LS BMC (P < 0.05). The interaction effect of muscular fitness showed a trend in the association of VEGF with TBLH BMC (P = 0.122) and TNF-α with LS BMC (P = 0.057). Stratified analyses by muscular fitness levels showed an inverse association of VEGF with TBLH BMC (β = -0.152) and TNF-α with LS BMC (β = -0.491) in the low-fitness group, while no association was found in the high-fitness group. CONCLUSION IL-6, VEGF, TNF-α, and IL-1β are significantly associated with bone mass. Higher muscular fitness may attenuate the adverse effect of high VEGF and TNF-α on bone mass.
Collapse
|
32
|
Khan YA, Maurya SK, Kulkarni C, Tiwari MC, Nagar GK, Chattopadhyay N. Fasciola
helminth defense molecule‐1 protects against experimental arthritis by inhibiting osteoclast formation and function without modulating the systemic immune response. FASEB J 2019; 34:1091-1106. [DOI: 10.1096/fj.201901480rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/05/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Yasir Akhtar Khan
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
- Section of Parasitology Department of Zoology Aligarh Muslim University Aligarh India
| | | | - Chirag Kulkarni
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
- Academy of Scientific and Innovative Research CSIR‐Central Drug Research Institute Lucknow India
| | | | - Geet Kumar Nagar
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
| | - Naibedya Chattopadhyay
- Division of Endocrinology CSIR‐Central Drug Research Institute Lucknow India
- Academy of Scientific and Innovative Research CSIR‐Central Drug Research Institute Lucknow India
| |
Collapse
|
33
|
Zhao C, Yu T, Dou Q, Guo Y, Yang X, Chen Y. Knockout of TLR4 promotes fracture healing by activating Wnt/β-catenin signaling pathway. Pathol Res Pract 2019; 216:152766. [PMID: 31796334 DOI: 10.1016/j.prp.2019.152766] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/13/2019] [Accepted: 11/25/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the effect of Toll like receptor 4 (TLR4) on fracture healing. METHODS The open tibial fracture models in TLR4 knockout (TLR4-/-) and wild type (WT) C57BL-6 J mice were established. The radiological examination, tartrate-resistant acid phosphatase (TRAP) staining, Micro-CT scan and biological torsion test were performed on 7, 14 and 21 days after operation. Enzyme Linked Immunosorbent Assay (ELISA) kit was used to detect the expression levels of tumor necrosis factor-α (TNF-α), interleukin-1 beta (IL-1β) and interleukin 6 (IL-6). Western blotting was used to detect the expression of β-catenin, Wingless-type MMTV integration site family, member 4 and 5B (Wnt4 and Wnt5B), proliferating cell nuclear antigen (PCNA) and bone morphogenetic protein-2 (BMP-2) of the callus tissue obtained from mice. RESULTS TLR4 knockout promoted fracture healing, reduced the number of osteoclasts, increased bone callus volume (BV) and callus mineralized volume fraction (BV/TV%) (P < 0.05), increased the maximum torque and torsional stiffness of callus (P < 0.05), reduced TNF-α, IL-1β and IL-6 expression (P < 0.01), and increased the expression levels of β-catenin, Wnt4, Wnt5B, PCNA and BMP-2 (P < 0.01). CONCLUSION TLR4 knockout reduced inflammatory and promoted fracture healing by activating Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Cunju Zhao
- Department of Spinal Surgery, Qilu Hospital of ShanDong University, No. 107, Cultural West Road, Jinan City, Shandong Province, 250012, China; Department Of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng City, Shandong Province, 252000, China
| | - Tao Yu
- Department of Spinal Surgery, Qilu Hospital of ShanDong University, No. 107, Cultural West Road, Jinan City, Shandong Province, 250012, China; Department Of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng City, Shandong Province, 252000, China
| | - Qingjun Dou
- Department Of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng City, Shandong Province, 252000, China
| | - Yue Guo
- Department Of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng City, Shandong Province, 252000, China
| | - Xiaofei Yang
- Department Of Orthopedics, Liaocheng People's Hospital, No. 67, Dongchang West Road, Liaocheng City, Shandong Province, 252000, China
| | - Yunzhen Chen
- Department of Spinal Surgery, Qilu Hospital of ShanDong University, No. 107, Cultural West Road, Jinan City, Shandong Province, 250012, China.
| |
Collapse
|
34
|
Orsolini G, Fassio A, Rossini M, Adami G, Giollo A, Caimmi C, Idolazzi L, Viapiana O, Gatti D. Effects of biological and targeted synthetic DMARDs on bone loss in rheumatoid arthritis. Pharmacol Res 2019; 147:104354. [DOI: 10.1016/j.phrs.2019.104354] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022]
|
35
|
KAWABATA T, TOKUDA H, FUJITA K, MATSUSHIMA-NISHIWAKI R, SAKAI G, TACHI J, HIOKI T, KIM W, IIDA H, OTSUKA T, KOZAWA O. HSP90 inhibitors diminish PDGF-BB-induced migration of osteoblasts via suppression of p44/p42 MAP kinase. Biomed Res 2019; 40:169-178. [DOI: 10.2220/biomedres.40.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Tetsu KAWABATA
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences
- Department of Pharmacology, Gifu University Graduate School of Medicine
- Department of Orthopedic Surgery, Toyokawa City Hospital
| | - Haruhiko TOKUDA
- Department of Pharmacology, Gifu University Graduate School of Medicine
- Department of Clinical Laboratory/Medical Genome Center Biobank, National Center for Geriatrics and Gerontology
| | - Kazuhiko FUJITA
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | | | - Go SAKAI
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Junko TACHI
- Department of Pharmacology, Gifu University Graduate School of Medicine
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Tomoyuki HIOKI
- Department of Pharmacology, Gifu University Graduate School of Medicine
- Department of Dermatology, Kizawa Memorial Hospital
| | - Woo KIM
- Department of Pharmacology, Gifu University Graduate School of Medicine
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Hiroki IIDA
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Takanobu OTSUKA
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences
| | - Osamu KOZAWA
- Department of Pharmacology, Gifu University Graduate School of Medicine
| |
Collapse
|
36
|
Metal Nanoparticles Released from Dental Implant Surfaces: Potential Contribution to Chronic Inflammation and Peri-Implant Bone Loss. MATERIALS 2019; 12:ma12122036. [PMID: 31242601 PMCID: PMC6630980 DOI: 10.3390/ma12122036] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
Abstract
Peri-implantitis is an inflammatory disease affecting tissues surrounding dental implants. Although it represents a common complication of dental implant treatments, the underlying mechanisms have not yet been fully described. The aim of this study is to identify the role of titanium nanoparticles released form the implants on the chronic inflammation and bone lysis in the surrounding tissue. We analyzed the in vitro effect of titanium (Ti) particle exposure on mesenchymal stem cells (MSCs) and fibroblasts (FU), evaluating cell proliferation by MTT test and the generation of reactive oxygen species (ROS). Subsequently, in vivo analysis of peri-implant Ti particle distribution, histological, and molecular analyses were performed. Ti particles led to a time-dependent decrease in cell viability and increase in ROS production in both MSCs and FU. Tissue analyses revealed presence of oxidative stress, high extracellular and intracellular Ti levels and imbalanced bone turnover. High expression of ZFP467 and the presence of adipose-like tissue suggested dysregulation of the MSC population; alterations in vessel morphology were identified. The results suggest that Ti particles may induce the production of high ROS levels, recruiting abnormal quantity of neutrophils able to produce high level of metalloproteinase. This induces the degradation of collagen fibers. These events may influence MSC commitment, with an imbalance of bone regeneration.
Collapse
|
37
|
Zhang T, Yao Y. Effects of inflammatory cytokines on bone/cartilage repair. J Cell Biochem 2019; 120:6841-6850. [PMID: 30335899 DOI: 10.1002/jcb.27953] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/02/2018] [Indexed: 01/24/2023]
Abstract
Many inflammatory factors can affect cell behaviors and work as a form of inter-regulatory networks through the inflammatory pathway. Inflammatory cytokines are critical for triggering bone regeneration after fracture or bone injury. Also, inflammatory cytokines play an important role in cartilage repair. The synergistic or antagonistic effects of both proinflammatory and anti-inflammatory cytokines have a great influence on fracture healing. This review discusses key inflammatory cytokines and signaling pathways involved in bone or cartilage repair.
Collapse
Affiliation(s)
- Tingshuai Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
The Emerging Role of Mesenchymal Stem Cells in Vascular Calcification. Stem Cells Int 2019; 2019:2875189. [PMID: 31065272 PMCID: PMC6466855 DOI: 10.1155/2019/2875189] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/12/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
Vascular calcification (VC), characterized by hydroxyapatite crystal depositing in the vessel wall, is a common pathological condition shared by many chronic diseases and an independent risk factor for cardiovascular events. Recently, VC is regarded as an active, dynamic cell-mediated process, during which calcifying cell transition is critical. Mesenchymal stem cells (MSCs), with a multidirectional differentiation ability and great potential for clinical application, play a duplex role in the VC process. MSCs facilitate VC mainly through osteogenic transformation and apoptosis. Meanwhile, several studies have reported the protective role of MSCs. Anti-inflammation, blockade of the BMP2 signal, downregulation of the Wnt signal, and antiapoptosis through paracrine signaling are possible mechanisms. This review displays the evidence both on the facilitating role and on the protective role of MSCs, then discusses the key factors determining this divergence.
Collapse
|
39
|
Hu K, Jiang W, Sun H, Li Z, Rong G, Yin Z. Long noncoding RNA ZBED3‐AS1 induces the differentiation of mesenchymal stem cells and enhances bone regeneration by repressing IL‐1β via Wnt/β‐catenin signaling pathway. J Cell Physiol 2019; 234:17863-17875. [DOI: 10.1002/jcp.28416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/01/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Kongzu Hu
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei Anhui P. R. China
| | - Wei Jiang
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei Anhui P. R. China
| | - Heyan Sun
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei Anhui P. R. China
| | - Zhenwei Li
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei Anhui P. R. China
| | - Genxiang Rong
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei Anhui P. R. China
| | - Zongsheng Yin
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei Anhui P. R. China
| |
Collapse
|
40
|
Guo YC, Chiu YH, Chen CP, Wang HS. Interleukin-1β induces CXCR3-mediated chemotaxis to promote umbilical cord mesenchymal stem cell transendothelial migration. Stem Cell Res Ther 2018; 9:281. [PMID: 30359318 PMCID: PMC6202827 DOI: 10.1186/s13287-018-1032-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are known to home to injured and inflamed regions via the bloodstream to assist in tissue regeneration in response to signals of cellular damage. However, the factors and mechanisms that affect their transendothelial migration are still unclear. In this study, the mechanisms involved in interleukin-1β (IL-1β) enhancing the transendothelial migration of MSCs were investigated. METHODS Immunofluorescence staining and Western blotting were used to observe IL-1β-induced CXC chemokine receptor 3 (CXCR3) expression on MSCs. Quantitative real-time PCR and ELISA were used to demonstrate IL-1β upregulated both chemokine (C-X-C motif) ligand 9 (CXCL9) mRNA and CXCL9 ligand secretion in human umbilical vein endothelial cells (HUVECs). Monolayer co-cultivation, agarose drop chemotaxis, and transwell assay were conducted to investigate the chemotaxis invasion and transendothelial migration ability of IL-1β-induced MSCs in response to CXCL9. RESULTS In this study, our immunofluorescence staining showed that IL-1β induces CXCR3 expression on MSCs. This result was confirmed by Western blotting. Following pretreatment with protein synthesis inhibitor cycloheximide, we found that IL-1β induced CXCR3 on the surface of MSCs via protein synthesis pathway. Quantitative real-time PCR and ELISA validated that IL-1β upregulated both CXCL9 mRNA and CXCL9 ligand secretion in HUVECs. In response to CXCL9, chemotaxis invasion and transendothelial migration ability were increased in IL-1β-stimulated MSCs. In addition, we pretreated MSCs with CXCR3 antagonist AMG-487 and p38 MAPK inhibitor SB203580 to confirm CXCR3-CXCL9 interaction and the role of CXCR3 in IL-1β-induced chemotaxis invasion and transendothelial migration. CONCLUSION We found that IL-1β induces the expression of CXCR3 through p38 MAPK signaling and that IL-1β also enhances CXCL9 ligand secretion in HUVECs. These results indicated that IL-1β promotes the transendothelial migration of MSCs through CXCR3-CXCL9 axis. The implication of the finding could enhance the efficacy of MSCs homing to target sites.
Collapse
Affiliation(s)
- Yu-Chien Guo
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming University, Peitou, Taipei, 112, Taiwan, Republic of China
| | - Yun-Hsuan Chiu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming University, Peitou, Taipei, 112, Taiwan, Republic of China
| | - Chie-Pein Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Hwai-Shi Wang
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming University, Peitou, Taipei, 112, Taiwan, Republic of China.
| |
Collapse
|
41
|
Jenei-Lanzl Z, Meurer A, Zaucke F. Interleukin-1β signaling in osteoarthritis - chondrocytes in focus. Cell Signal 2018; 53:212-223. [PMID: 30312659 DOI: 10.1016/j.cellsig.2018.10.005] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/07/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) can be regarded as a chronic, painful and degenerative disease that affects all tissues of a joint and one of the major endpoints being loss of articular cartilage. In most cases, OA is associated with a variable degree of synovial inflammation. A variety of different cell types including chondrocytes, synovial fibroblasts, adipocytes, osteoblasts and osteoclasts as well as stem and immune cells are involved in catabolic and inflammatory processes but also in attempts to counteract the cartilage loss. At the molecular level, these changes are regulated by a complex network of proteolytic enzymes, chemokines and cytokines (for review: [1]). Here, interleukin-1 signaling (IL-1) plays a central role and its effects on the different cell types involved in OA are discussed in this review with a special focus on the chondrocyte.
Collapse
Affiliation(s)
- Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt/Main, Germany
| | - Andrea Meurer
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt/Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt/Main, Germany.
| |
Collapse
|
42
|
Bone erosions in rheumatoid arthritis: recent developments in pathogenesis and therapeutic implications. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2018; 18:304-319. [PMID: 30179207 PMCID: PMC6146189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Bone erosions develop early in the course of rheumatoid arthritis (RA) and deteriorate progressively, causing joint damage and resulting in impaired functional capacity of patients. During the last years, considerable number of studies has increased our understanding of the pathogenetic mechanisms mediating the development of bone erosions in RA. Increased production of RANKL and other cytokines, dysregulation of innate immune mechanisms, autoantibodies specific to RA and alterations of microRNA expression stimulate differentiation and function of osteoclasts, which are responsible for the development of bone erosions. Besides, increased levels of cytokines, overproduction of antagonists of the canonical Wnt signaling pathway and deficient production of bone morphogenetic proteins result in impaired osteoblast differentiation and function, undermining the capacity of bone erosions to repair. Disease-modifying antirheumatic drugs, synthetic or biological, currently used in the treatment of RA, can halt the progression of bone erosions and may even lead to partial repair, although complete repair is unattainable. Targeting pathogenetic mechanisms participating in the erosive process may add to the therapeutic effect of DMARDs and help in the prevention or repair of bone erosions. However, more studies are still needed to confirm whether such therapeutic strategies are effective.
Collapse
|
43
|
Liu Q, Zhou Y, Li Z. PDGF‑BB promotes the differentiation and proliferation of MC3T3‑E1 cells through the Src/JAK2 signaling pathway. Mol Med Rep 2018; 18:3719-3726. [PMID: 30106097 PMCID: PMC6131220 DOI: 10.3892/mmr.2018.9351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 07/20/2018] [Indexed: 11/20/2022] Open
Abstract
Platelet-derived growth factor-BB (PDGF-BB) serves a critical function in human osteoblast differentiation and proliferation. Src and Janus kinase 2 (JAK2) are involved in these processes. In our previous study, it was identified that Src could promote the phosphorylation of JAK2. However, it has yet to be determined whether the Src/JAK2 signaling pathway affects PDGF-BB-mediated osteoblast differentiation and proliferation. In the present study, western blotting, polymerase chain reaction, alizarin red staining, alkaline phosphatase and Cell Counting kit-8 were employed to explore these questions. Firstly, it was demonstrated that PDGF-BB activates the Src/JAK2 signaling pathway in MC3T3-E1 cells in a time-dependent manner. Furthermore, it was demonstrated that PDGF-BB expression promoted MC3T3-E1 cell differentiation and proliferation; this process was suppressed by AG1295, SU6656 and AG490, which are inhibitors of PDGFR-β, Src and JAK2, respectively. SU6656 downregulated the activity of Src and JAK2, while AG490 only downregulated JAK2 activity. Therefore, it was concluded that Src is upstream of JAK2. PDGF-BB also upregulated the expression of osteogenesis-associated genes, and the formation of mineral nodules. However, these effects were markedly inhibited by treatment with SU6656. This indicated that PDGF-BB promoted MC3T3-E1 cell differentiation and proliferation by activating the Src/JAK2 signaling pathway. These results suggested that PDGF-BB may have potential applications in the treatment of osteoporosis and bone fractures.
Collapse
Affiliation(s)
- Qi Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Yunfeng Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‑MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, Hubei 430079, P.R. China
| | - Zubing Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
44
|
Fuggle NR, Westbury LD, Syddall HE, Duggal NA, Shaw SC, Maslin K, Dennison EM, Lord J, Cooper C. Relationships between markers of inflammation and bone density: findings from the Hertfordshire Cohort Study. Osteoporos Int 2018; 29:1581-1589. [PMID: 29808230 PMCID: PMC6093277 DOI: 10.1007/s00198-018-4503-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
UNLABELLED Among 365 Hertfordshire Cohort Study participants (aged 59-71 years at baseline), higher adiponectin and adiponectin to leptin ratios were associated with lower baseline lumbar spine and femoral neck bone mineral density (BMD). Lower IL-10 was associated with accelerated decline in lumbar spine BMD. This suggests that bone health can be influenced by changes in immune phenotype and alterations in adipokine homeostasis. INTRODUCTION The aim of this study was to examine the association between indices of inflammation and BMD in a population-based cohort of older adults in the UK. METHODS Analyses were based on a sample of 194 men and 171 women of the Hertfordshire Cohort Study (community-living, older adults). Dual energy X-ray absorptiometry (DXA) was performed at the lumbar spine and proximal femur at baseline and repeated at a median of 4.5 years (inter-quartile range 3.6 to 5.2). Inflammatory markers (CRP, TNF, IL-1β, IL-6, IL-8, IL-10, adiponectin and leptin) were ascertained at baseline using enzyme-linked immunosorbent assay (ELISA) techniques and Bio-Plex Pro Assays. Gender-adjusted linear regression was used to examine the associations between markers of inflammation and outcomes with and without adjustment for anthropometric and lifestyle factors. RESULTS The mean (SD) ages at baseline were 64.4 (2.5) and 66.5 (2.7) years for men and women respectively. Higher levels of adiponectin and adiponectin to leptin ratios were each associated with lower baseline lumbar spine and femoral neck BMD in gender-adjusted (p < 0.01) and fully adjusted (p < 0.05) analyses. Lower levels of IL-10 and TNF were each associated with accelerated decline in lumbar spine BMD in both gender-adjusted (p ≤ 0.05) and fully adjusted (p < 0.05) analyses. CONCLUSIONS In a cohort of older adults, high levels of adiponectin and adiponectin to leptin ratios were both associated with lower BMD at the lumbar spine and femoral neck at baseline, and lower IL-10 was associated with accelerated decline in BMD at the lumbar spine. This adds weight to the theory that bone health can be influenced by changes in immune phenotype and alterations in adipokine homeostasis.
Collapse
Affiliation(s)
- N R Fuggle
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - L D Westbury
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - H E Syddall
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - N A Duggal
- MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - S C Shaw
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - K Maslin
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - E M Dennison
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
- Victoria University of Wellington, Wellington, New Zealand
| | - J Lord
- MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - C Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK.
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
- NIHR Musculoskeletal Biomedical Research Unit, University of Oxford, Oxford, UK.
| |
Collapse
|
45
|
Movilla N, Borau C, Valero C, García-Aznar JM. Degradation of extracellular matrix regulates osteoblast migration: A microfluidic-based study. Bone 2018; 107:10-17. [PMID: 29107125 DOI: 10.1016/j.bone.2017.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 01/06/2023]
Abstract
Bone regeneration is strongly dependent on the capacity of cells to move in a 3D microenvironment, where a large cascade of signals is activated. To improve the understanding of this complex process and to advance in the knowledge of the role of each specific signal, it is fundamental to analyze the impact of each factor independently. Microfluidic-based cell culture is an appropriate technology to achieve this objective, because it allows recreating realistic 3D local microenvironments by taking into account the extracellular matrix, cells and chemical gradients in an independent or combined scenario. The main aim of this work is to analyze the impact of extracellular matrix properties and growth factor gradients on 3D osteoblast movement, as well as the role of cell matrix degradation. For that, we used collagen-based hydrogels, with and without crosslinkers, under different chemical gradients, and eventually inhibiting metalloproteinases to tweak matrix degradation. We found that osteoblast's 3D migratory patterns were affected by the hydrogel properties and the PDGF-BB gradient, although the strongest regulatory factor was determined by the ability of cells to remodel the matrix.
Collapse
Affiliation(s)
- N Movilla
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain
| | - C Borau
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain
| | - C Valero
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain
| | - J M García-Aznar
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain.
| |
Collapse
|
46
|
Turner JD, Naylor AJ, Buckley C, Filer A, Tak PP. Fibroblasts and Osteoblasts in Inflammation and Bone Damage. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:37-54. [DOI: 10.1007/978-3-319-78127-3_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
47
|
Hoff P, Gaber T, Strehl C, Schmidt-Bleek K, Lang A, Huscher D, Burmester GR, Schmidmaier G, Perka C, Duda GN, Buttgereit F. Immunological characterization of the early human fracture hematoma. Immunol Res 2017; 64:1195-1206. [PMID: 27629117 DOI: 10.1007/s12026-016-8868-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The initial inflammatory phase of fracture healing is of great importance for the clinical outcome. We aimed to develop a detailed time-dependent analysis of the initial fracture hematoma. We analyzed the composition of immune cell subpopulations by flow cytometry and the concentration of cytokines and chemokines by bioplex in 42 samples from human fractures of long bones <72 h post-trauma. The early human fracture hematoma is characterized by maturation of granulocytes and migration of monocytes/macrophages and hematopoietic stem cells. Both T helper cells and cytotoxic T cells proliferate within the fracture hematoma and/or migrate to the fracture site. Humoral immunity characteristics comprise high concentration of pro-inflammatory cytokines such as IL-6, IL-8, IFNγ and TNFα, but also elevated concentration of anti-inflammatory cytokines, e.g., IL-1 receptor antagonist and IL-10. Furthermore, we found that cells of the fracture hematoma represent a source for key chemokines. Even under the bioenergetically restricted conditions that exist in the initial fracture hematoma, immune cells are not only present, but also survive, mature, function and migrate. They secrete a cytokine/chemokine cocktail that contributes to the onset of regeneration. We hypothesize that this specific microenvironment of the initial fracture hematoma is among the crucial factors that determine fracture healing.
Collapse
Affiliation(s)
- Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany.
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany.
| | - T Gaber
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| | - C Strehl
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
| | - K Schmidt-Bleek
- Julius Wolff Institute, Charité University Hospital, 13353, Berlin, Germany
| | - A Lang
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353, Berlin, Germany
| | - D Huscher
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
| | - G R Burmester
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
| | - G Schmidmaier
- Department of Orthopedics, University Hospital Heidelberg, 69118, Heidelberg, Germany
| | - C Perka
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
- Center for Musculoskeletal Surgery, Charité University Hospital, 10117, Berlin, Germany
| | - G N Duda
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
- Julius Wolff Institute, Charité University Hospital, 13353, Berlin, Germany
| | - F Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Charitéplatz 1, 10117, Berlin, Germany
- German Arthritis Research Center (DRFZ), 10117, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| |
Collapse
|
48
|
Kainuma S, Tokuda H, Yamamoto N, Kuroyanagi G, Fujita K, Kawabata T, Sakai G, Matsushima-Nishiwaki R, Kozawa O, Otsuka T. Heat shock protein 27 (HSPB1) suppresses the PDGF-BB-induced migration of osteoblasts. Int J Mol Med 2017; 40:1057-1066. [PMID: 28902366 PMCID: PMC5593454 DOI: 10.3892/ijmm.2017.3119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022] Open
Abstract
Heat shock protein 27 (HSP27/HSPB1), one of the small heat shock proteins, is constitutively expressed in various tissues. HSP27 and its phosphorylation state participate in the regulation of multiple physiological and pathophysiological cell functions. However, the exact roles of HSP27 in osteoblasts remain unclear. In the present study, we investigated the role of HSP27 in the platelet-derived growth factor‑BB (PDGF‑BB)‑stimulated migration of osteoblast-like MC3T3-E1 cells. PDGF-BB by itself barely upregulated the expression of HSP27 protein, but stimulated the phosphorylation of HSP27 in these cells. The PDGF-BB‑induced cell migration was significantly downregulated by HSP27 overexpression. The PDGF-BB-induced migrated cell numbers of the wild‑type HSP27-overexpressing cells and the phospho‑mimic HSP27-overexpressing (3D) cells were less than those of the unphosphorylatable HSP27-overexpressing (3A) cells. PD98059, an inhibitor of MEK1/2, SB203580, an inhibitor of p38 mitogen-activated protein kinase, and SP600125, an inhibitor of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) reduced the PDGF-BB-induced migration of these cells, whereas Akt inhibitor or rapamycin, an inhibitor of upstream kinase of p70 S6 kinase (mTOR), barely affected the migration. However, the PDGF-BB-induced phosphorylation of p44/p42 MAPΚ, p38 MAPK and SAPK/JNK was not affected by HSP27 overexpression. There were no significant differences in the phosphorylation of p44/p42 MAPΚ, p38 MAP kinase or SAPK/JNK between the 3D cells and the 3A cells. These results strongly suggest that HSP27 functions as a negative regulator in the PDGF-BB-stimulated migration of osteoblasts, and the suppressive effect is amplified by the phosphorylation state of HSP27.
Collapse
Affiliation(s)
- Shingo Kainuma
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Naohiro Yamamoto
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Gen Kuroyanagi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Kazuhiko Fujita
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Tetsu Kawabata
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Go Sakai
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
49
|
Wehmeyer C, Pap T, Buckley CD, Naylor AJ. The role of stromal cells in inflammatory bone loss. Clin Exp Immunol 2017; 189:1-11. [PMID: 28419440 PMCID: PMC5461090 DOI: 10.1111/cei.12979] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2017] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation, local and systemic bone loss and a lack of compensatory bone repair. Fibroblast-like synoviocytes (FLS) are the most abundant cells of the stroma and a key population in autoimmune diseases such as RA. An increasing body of evidence suggests that these cells play not only an important role in chronic inflammation and synovial hyperplasia, but also impact bone remodelling. Under inflammatory conditions FLS release inflammatory cytokines, regulate bone destruction and formation and communicate with immune cells to control bone homeostasis. Other stromal cells, such as osteoblasts and terminally differentiated osteoblasts, termed osteocytes, are also involved in the regulation of bone homeostasis and are dysregulated during inflammation. This review highlights our current understanding of how stromal cells influence the balance between bone formation and bone destruction. Increasing our understanding of these processes is critical to enable the development of novel therapeutic strategies with which to treat bone loss in RA.
Collapse
Affiliation(s)
- C. Wehmeyer
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth HospitalBirminghamUK
| | - T. Pap
- Institute of Experimental Musculoskeletal Medicine, University Hospital MuensterMuensterGermany
| | - C. D. Buckley
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth HospitalBirminghamUK
| | - A. J. Naylor
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth HospitalBirminghamUK
| |
Collapse
|
50
|
Thiel A, Reumann MK, Boskey A, Wischmann J, von Eisenhart-Rothe R, Mayer-Kuckuk P. Osteoblast migration in vertebrate bone. Biol Rev Camb Philos Soc 2017. [PMID: 28631442 DOI: 10.1111/brv.12345] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bone formation, for example during bone remodelling or fracture repair, requires mature osteoblasts to deposit bone with remarkable spatial precision. As osteoblast precursors derive either from circulation or resident stem cell pools, they and their progeny are required to migrate within the three-dimensional bone space and to navigate to their destination, i.e. to the site of bone formation. An understanding of this process is emerging based on in vitro and in vivo studies of several vertebrate species. Receptors on the osteoblast surface mediate cell adhesion and polarization, which induces osteoblast migration. Osteoblast migration is then facilitated along gradients of chemoattractants. The latter are secreted or released proteolytically by several cell types interacting with osteoblasts, including osteoclasts and vascular endothelial cells. The positions of these cellular sources of chemoattractants in relation to the position of the osteoblasts provide the migrating osteoblasts with tracks to their destination, and osteoblasts possess the means to follow a track marked by multiple chemoattractant gradients. In addition to chemotactic cues, osteoblasts sense other classes of signals and utilize them as landmarks for navigation. The composition of the osseous surface guides adhesion and hence migration efficiency and can also provide steering through haptotaxis. Further, it is likely that signals received from surface interactions modulate chemotaxis. Besides the nature of the surface, mechanical signals such as fluid flow may also serve as navigation signals for osteoblasts. Alterations in osteoblast migration and navigation might play a role in metabolic bone diseases such as osteoporosis.
Collapse
Affiliation(s)
- Antonia Thiel
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| | - Marie K Reumann
- Siegfried Weller Institute, BG Hospital, University of Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Adele Boskey
- Mineralized Tissue Laboratory, Research Division, Hospital for Special Surgery, 535 E 70th Street, New York, NY 10021, U.S.A
| | - Johannes Wischmann
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| | - Rüdiger von Eisenhart-Rothe
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| | - Philipp Mayer-Kuckuk
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| |
Collapse
|