1
|
Chen H, Lin H, Dai J. Causal associations of interleukins and osteoporosis: A genetic correlation study. Medicine (Baltimore) 2024; 103:e39036. [PMID: 39121248 PMCID: PMC11315551 DOI: 10.1097/md.0000000000039036] [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: 10/19/2023] [Accepted: 07/01/2024] [Indexed: 08/11/2024] Open
Abstract
The association between interleukins and osteoporosis has attracted much attention these days. However, the causal relationship between them is uncertain. Hence, this study performed a Mendelian randomization (MR) analysis to investigate the causal effects of interleukins on osteoporosis. The summary data for interleukins and osteoporosis came from 4 different genome-wide association studies. Significant and independent (P < 5 × 10-6; r2 < 0.001, 10,000 kbp) single-nucleotide polymorphisms were extracted for MR analysis. The inverse-variance weighted and other methods were used for MR analysis, while sensitivity analyses were conducted to test the reliability and stability. The positive causal effects of interleukin-7 on osteoporosis (odds ratio = 1.084; 95% confidence interval: 1.010-1.163; P = .025) were observed. No causal relationship was found between other interleukins and osteoporosis. In the sensitivity analysis, the results did not show the presence of pleiotropy and heterogeneity. Therefore, the results were robust for the MR analysis. This study revealed that interleukin-7 was positively related to osteoporosis and that other interleukins were not related to osteoporosis.
Collapse
Affiliation(s)
- Huihuang Chen
- Emergency Department, The Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Haibin Lin
- Department of Orthopedics, The Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Jianhui Dai
- Department of Orthopedics, The Affiliated Hospital of Putian University, Putian, Fujian Province, China
| |
Collapse
|
2
|
Hong J, Luo F, Du X, Xian F, Li X. The immune cells in modulating osteoclast formation and bone metabolism. Int Immunopharmacol 2024; 133:112151. [PMID: 38685175 DOI: 10.1016/j.intimp.2024.112151] [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: 01/07/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Osteoclasts are pivotal in regulating bone metabolism, with immune cells significantly influencing both physiological and pathological processes by modulating osteoclast functions. This is particularly evident in conditions of inflammatory bone resorption, such as rheumatoid arthritis and periodontitis. This review summarizes and comprehensively analyzes the research progress on the regulation of osteoclast formation by immune cells, aiming to unveil the underlying mechanisms and pathways through which diseases, such as rheumatoid arthritis and periodontitis, impact bone metabolism.
Collapse
Affiliation(s)
- Jiale Hong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Fang Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xingyue Du
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Fa Xian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
3
|
Mou K, Chan SMH, Vlahos R. Musculoskeletal crosstalk in chronic obstructive pulmonary disease and comorbidities: Emerging roles and therapeutic potentials. Pharmacol Ther 2024; 257:108635. [PMID: 38508342 DOI: 10.1016/j.pharmthera.2024.108635] [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: 11/06/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a multifaceted respiratory disorder characterized by progressive airflow limitation and systemic implications. It has become increasingly apparent that COPD exerts its influence far beyond the respiratory system, extending its impact to various organ systems. Among these, the musculoskeletal system emerges as a central player in both the pathogenesis and management of COPD and its associated comorbidities. Muscle dysfunction and osteoporosis are prevalent musculoskeletal disorders in COPD patients, leading to a substantial decline in exercise capacity and overall health. These manifestations are influenced by systemic inflammation, oxidative stress, and hormonal imbalances, all hallmarks of COPD. Recent research has uncovered an intricate interplay between COPD and musculoskeletal comorbidities, suggesting that muscle and bone tissues may cross-communicate through the release of signalling molecules, known as "myokines" and "osteokines". We explored this dynamic relationship, with a particular focus on the role of the immune system in mediating the cross-communication between muscle and bone in COPD. Moreover, we delved into existing and emerging therapeutic strategies for managing musculoskeletal disorders in COPD. It underscores the development of personalized treatment approaches that target both the respiratory and musculoskeletal aspects of COPD, offering the promise of improved well-being and quality of life for individuals grappling with this complex condition. This comprehensive review underscores the significance of recognizing the profound impact of COPD on the musculoskeletal system and its comorbidities. By unravelling the intricate connections between these systems and exploring innovative treatment avenues, we can aspire to enhance the overall care and outcomes for COPD patients, ultimately offering hope for improved health and well-being.
Collapse
Affiliation(s)
- Kevin Mou
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Stanley M H Chan
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
4
|
Ballikaya E, Babadag S, San Keskin NO, Çelebi-Saltik B. The Effects of Grape Seed Oligomeric Proanthocyanidin and Nisin on Dental Pulp Stem Cells. Acta Stomatol Croat 2024; 58:2-17. [PMID: 38562220 PMCID: PMC10981911 DOI: 10.15644/asc58/1/1] [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: 10/25/2023] [Accepted: 01/09/2024] [Indexed: 04/04/2024] Open
Abstract
Objective This study aimed to evaluate the biological effects of "proanthocyanidin" (PA), and "nisin" (Ni), on dental pulp stem cells (DPSCs) and LPS-induced DPSCs as well as their antimicrobial effects against S. aureus and E. coli. Materials and methods After characterization of DPSCs, cytotoxicity of PA and Ni on DPSCs were evaluated using a water-soluble tetrazolium salt (WST-1). The cytokines and chemokines released by DPSCs and the expression levels of IL-6, IL-8, and TNF alpha were detected with human Cytokine Array C5 and enzyme-linked immunosorbent assay (ELİSA), respectively. The antibacterial activities of PA and Ni were tested using the drop plate method. Results PA at 75 μg/ml increased cell viability, decreased TNF-α expression of DPSCs, did not show any cytotoxic effects on LPS-induced DPSCs, and also showed a tendency to decrease TNF-α expression. PA at 75 μg/ml exhibited higher expressions of TIMP-2, OPG, IL-7, and IL-8 in LPS-induced DPSCs compared to DPSCs. Ni at 100 μg/ml decreased TNF-α expression in DPSCs with no cytotoxic effects. It provided increased cell viability and a downregulation trend of TNF-α expression in LPS-induced DPSCs. Both Ni and PA provided strong antibacterial effects against S. aureus. Ni at 200μg/ml had strong antibacterial effects against E. coli without affecting negatively the viability of both DPSCs and LPS-induced DPSCs and showed anti-inflammatory activity by decreasing TNF-α expression. PA provided strong antibacterial effects against E. coli at 200 μg/ml but affected DPSCs viability negatively. Conclusion PA and Ni at specific concentrations exhibited immunomodulatory activity on DPSCs and LPS-induced DPSCs without any cytotoxic effects and strong antibacterial effects on S. aureus.
Collapse
Affiliation(s)
- Elif Ballikaya
- Department of Oral and Dental Health Research, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Department of Pediatric Dentistry, Hacettepe University Faculty of Dentistry, Ankara, Turkey
| | - Sena Babadag
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Nalan Oya San Keskin
- Polatlı Science and Literature Faculty, Nanosan Laboratory, Ankara Hacı Bayram Veli University, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Oral and Dental Health Research, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
5
|
Xiao H, Wang Y, Wang Z, Wang B, Hu L, Hou J, Du K, Sun N, Wang L. Angelica sinensis polysaccharides ameliorated 5-Fluorouracil-induced damage of early B cell progenitors by alleviating oxidative stress of IL-7 producing mesenchymal stem and progenitor cells. Biomed Pharmacother 2023; 167:115599. [PMID: 37783150 DOI: 10.1016/j.biopha.2023.115599] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
B-lymphocytopenia among myelosuppression is the most intractable side effect of chemotherapy. Here, we investigated ways to alleviate 5-fluorouracil-caused stress hematopoietic impairment. We found that intraperitoneally injected ASP (Angelica sinensis polysaccharides) (100 mg/kg per day), one main active ingredient of Angelica sinensis, for consecutive 7 days, significantly recovered mouse bone marrow pro-B and pre-B cells, reversed the capacity of CFU-PreB colony forming, thus alleviating B cell reduction in the spleen and peripheral blood, as well as ameliorating immunoglobin from spleen and serum. The mechanism is related to the protective effects of ASP on IL-7 producing cells, including perivascular Leptin+ and CXCL12+ mesenchymal stem and progenitor cells (MSPCs), thus promoting IL-7 production, and activating IL-7R-mediated STAT5, PI3K-AKT signaling, including survival signals and EBF1, PAX5 transcription factor expression. Additionally, ASP's IL-7 promoting effect was demonstrated to be associated with maintaining osteogenesis/adipogenesis balance of MSPCs via the NRF2 antioxidant pathway. Collectively, our findings indicate that ASP reverse stress B-lymphocytopenia via improving Nrf2 signaling, promoting IL-7 production in MSPCs, and subsequently maintaining survival, proliferation, and differentiation of B cell progenitors, which may represent a promising therapeutic strategy.
Collapse
Affiliation(s)
- Hanxianzhi Xiao
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China; Chongqing Blood Center, Chongqing 400015, China
| | - Yaping Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ziling Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Biyao Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ling Hu
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Jiying Hou
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Kunhang Du
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Nianci Sun
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Lu Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
6
|
Tang J, Yang L, Guan F, Miller H, Camara NOS, James LK, Benlagha K, Kubo M, Heegaard S, Lee P, Lei J, Zeng H, He C, Zhai Z, Liu C. The role of Raptor in lymphocytes differentiation and function. Front Immunol 2023; 14:1146628. [PMID: 37283744 PMCID: PMC10239924 DOI: 10.3389/fimmu.2023.1146628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/28/2023] [Indexed: 06/08/2023] Open
Abstract
Raptor, a key component of mTORC1, is required for recruiting substrates to mTORC1 and contributing to its subcellular localization. Raptor has a highly conserved N-terminus domain and seven WD40 repeats, which interact with mTOR and other mTORC1-related proteins. mTORC1 participates in various cellular events and mediates differentiation and metabolism. Directly or indirectly, many factors mediate the differentiation and function of lymphocytes that is essential for immunity. In this review, we summarize the role of Raptor in lymphocytes differentiation and function, whereby Raptor mediates the secretion of cytokines to induce early lymphocyte metabolism, development, proliferation and migration. Additionally, Raptor regulates the function of lymphocytes by regulating their steady-state maintenance and activation.
Collapse
Affiliation(s)
- Jianing Tang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Louisa K. James
- Centre for Immunobiology, Bizard Institute, Queen Mary University of London, London, United Kingdom
| | - Kamel Benlagha
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, Paris, France
| | - Masato Kubo
- Laboratory for Cytokine Regulation, Center for Integrative Medical Science (IMS), Rikagaku Kenkyusho, Institute of Physical and Chemical Research (RIKEN) Yokohama Institute, Yokohama, Japan
| | - Steffen Heegaard
- Department of Ophthalmology, Rigshospitalet Glostrup, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hu Zeng
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
7
|
Grčević D, Sanjay A, Lorenzo J. Interactions of B-lymphocytes and bone cells in health and disease. Bone 2023; 168:116296. [PMID: 34942359 PMCID: PMC9936888 DOI: 10.1016/j.bone.2021.116296] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/09/2023]
Abstract
Bone remodeling occurs through the interactions of three major cell lineages, osteoblasts, which mediate bone formation, osteocytes, which derive from osteoblasts, sense mechanical force and direct bone turnover, and osteoclasts, which mediate bone resorption. However, multiple additional cell types within the bone marrow, including macrophages, T lymphocytes and B lymphocytes influence the process. The bone marrow microenvironment, which is supported, in part, by bone cells, forms a nurturing network for B lymphopoiesis. In turn, developing B lymphocytes influence bone cells. Bone health during homeostasis depends on the normal interactions of bone cells with other lineages in the bone marrow. In disease state these interactions become pathologic and can cause abnormal function of bone cells and inadequate repair of bone after a fracture. This review summarizes what is known about the development of B lymphocytes and the interactions of B lymphocytes with bone cells in both health and disease.
Collapse
Affiliation(s)
- Danka Grčević
- Department of Physiology and Immunology, Croatian Institute for Brain Research, School of Medicine University of Zagreb, Zagreb, Croatia.
| | - Archana Sanjay
- Department of Orthopaedics, UConn Health, Farmington, CT, USA.
| | - Joseph Lorenzo
- Departments of Medicine and Orthopaedics, UConn Health, Farmington, CT, USA.
| |
Collapse
|
8
|
Estrogen-mediated downregulation of HIF-1α signaling in B lymphocytes influences postmenopausal bone loss. Bone Res 2022; 10:15. [PMID: 35177582 PMCID: PMC8854586 DOI: 10.1038/s41413-022-00189-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/18/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
In the bone marrow, B cells and bone-resorbing osteoclasts colocalize and form a specific microenvironment. How B cells functionally influence osteoclasts and bone architecture is poorly understood. Using genetically modified mice and high-throughput analyses, we demonstrate that prolonged HIF-1α signaling in B cells leads to enhanced RANKL production and osteoclast formation. In addition, deletion of HIF-1α in B cells prevents estrogen deficiency-induced bone loss in mice. Mechanistically, estrogen controls HIF-1α protein stabilization through HSP70-mediated degradation in bone marrow B cells. The stabilization of HIF-1α protein in HSP70-deficient bone marrow B cells promotes RANKL production and osteoclastogenesis. Induction of HSP70 expression by geranylgeranylacetone (GGA) administration alleviates ovariectomy-induced osteoporosis. Moreover, RANKL gene expression has a positive correlation with HIF1A expression in human B cells. In conclusion, HIF-1α signaling in B cells is crucial for the control of osteoclastogenesis, and the HSP70/HIF-1α axis may serve as a new therapeutic target for osteoporosis.
Collapse
|
9
|
Dissociation of Bone Resorption and Formation in Spaceflight and Simulated Microgravity: Potential Role of Myokines and Osteokines? Biomedicines 2022; 10:biomedicines10020342. [PMID: 35203551 PMCID: PMC8961781 DOI: 10.3390/biomedicines10020342] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
The dissociation of bone formation and resorption is an important physiological process during spaceflight. It also occurs during local skeletal unloading or immobilization, such as in people with neuromuscular disorders or those who are on bed rest. Under these conditions, the physiological systems of the human body are perturbed down to the cellular level. Through the absence of mechanical stimuli, the musculoskeletal system and, predominantly, the postural skeletal muscles are largely affected. Despite in-flight exercise countermeasures, muscle wasting and bone loss occur, which are associated with spaceflight duration. Nevertheless, countermeasures can be effective, especially by preventing muscle wasting to rescue both postural and dynamic as well as muscle performance. Thus far, it is largely unknown how changes in bone microarchitecture evolve over the long term in the absence of a gravity vector and whether bone loss incurred in space or following the return to the Earth fully recovers or partly persists. In this review, we highlight the different mechanisms and factors that regulate the humoral crosstalk between the muscle and the bone. Further we focus on the interplay between currently known myokines and osteokines and their mutual regulation.
Collapse
|
10
|
Gong Y, Yang J, Li X, Zhou C, Chen Y, Wang Z, Qiu X, Liu Y, Zhang H, Greenbaum J, Cheng L, Hu Y, Xie J, Yang X, Li Y, Bai Y, Wang YP, Chen Y, Tan LJ, Shen H, Xiao HM, Deng HW. A systematic dissection of human primary osteoblasts in vivo at single-cell resolution. Aging (Albany NY) 2021; 13:20629-20650. [PMID: 34428745 PMCID: PMC8436943 DOI: 10.18632/aging.203452] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022]
Abstract
Human osteoblasts are multifunctional bone cells, which play essential roles in bone formation, angiogenesis regulation, as well as maintenance of hematopoiesis. However, the categorization of primary osteoblast subtypes in vivo in humans has not yet been achieved. Here, we used single-cell RNA sequencing (scRNA-seq) to perform a systematic cellular taxonomy dissection of freshly isolated human osteoblasts from one 31-year-old male with osteoarthritis and osteopenia after hip replacement. Based on the gene expression patterns and cell lineage reconstruction, we identified three distinct cell clusters including preosteoblasts, mature osteoblasts, and an undetermined rare osteoblast subpopulation. This novel subtype was found to be the major source of the nuclear receptor subfamily 4 group A member 1 and 2 (NR4A1 and NR4A2) in primary osteoblasts, and the expression of NR4A1 was confirmed by immunofluorescence staining on mouse osteoblasts in vivo. Trajectory inference analysis suggested that the undetermined cluster, together with the preosteoblasts, are involved in the regulation of osteoblastogenesis and also give rise to mature osteoblasts. Investigation of the biological processes and signaling pathways enriched in each subpopulation revealed that in addition to bone formation, preosteoblasts and undetermined osteoblasts may also regulate both angiogenesis and hemopoiesis. Finally, we demonstrated that there are systematic differences between the transcriptional profiles of human and mouse osteoblasts, highlighting the necessity for studying bone physiological processes in humans rather than solely relying on mouse models. Our findings provide novel insights into the cellular heterogeneity and potential biological functions of human primary osteoblasts at the single-cell level.
Collapse
MESH Headings
- Adult
- Animals
- Cell Differentiation
- Cells, Cultured
- Humans
- Male
- Mice
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Osteoblasts/cytology
- Osteoblasts/metabolism
- Sequence Analysis, RNA
- Single-Cell Analysis
Collapse
Affiliation(s)
- Yun Gong
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Junxiao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaohua Li
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Cui Zhou
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yu Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Zun Wang
- Xiangya Nursing School, Central South University, Changsha 410013, China
| | - Xiang Qiu
- School of Basic Medical Science, Central South University, Changsha 410008, China
| | - Ying Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Huixi Zhang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Jonathan Greenbaum
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Liang Cheng
- Department of Orthopedics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuecheng Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuntong Bai
- Tulane Center for Bioinformatics and Genomics, Department of Biomedical Engineering, Tulane University, New Orleans, LA 70112, USA
| | - Yu-Ping Wang
- Tulane Center for Bioinformatics and Genomics, Department of Biomedical Engineering, Tulane University, New Orleans, LA 70112, USA
| | - Yiping Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA 70112, USA
| | - Li-Jun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hong-Mei Xiao
- Center of Reproductive Health, System Biology and Data Information, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha 410081, China
- School of Basic Medical Science, Central South University, Changsha 410008, China
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
- School of Basic Medical Science, Central South University, Changsha 410008, China
| |
Collapse
|
11
|
Making Sense of the Highly Variable Effects of Alcohol on Bone. Clin Rev Bone Miner Metab 2021. [DOI: 10.1007/s12018-021-09277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
12
|
Xu H, Cai L, Li Z, Zhang L, Wang G, Xie R, Jiang Y, Yuan Y, Nie H. Dual effect of IL-7/IL-7R signalling on the osteoimmunological system: a potential therapeutic target for rheumatoid arthritis. Immunology 2021; 164:161-172. [PMID: 33934341 DOI: 10.1111/imm.13351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/08/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
The IL-7/IL-7R pathway plays a vital role in the immune system, especially in the inflammatory response. Monocytes/macrophages (osteoclast precursors) have been recently recognized as important participants in the osteoclastogenesis of rheumatoid arthritis (RA) patients. Here, we aimed to investigate the therapeutic potential of IL-7/IL-7R pathway in RA and to determine whether it could restrain osteoclastogenic functions and therefore ameliorate RA. Firstly, collagen-induced arthritis (CIA) mice were administered with IL-7Rα-target antibodies to assess their therapeutic effect on arthritis. We found that blockade of the IL-7/IL-7R pathway protected CIA mice from bone destruction in addition to inducing inflammatory remission, by altering the RANKL/RANK/OPG ratio and consequently decreasing osteoclast formation. To explore the effect and mechanism of this pathway, bone marrow cells were induced to osteoclasts and treated with IL-7, a STAT5 inhibitor or supernatants from T cells. The results showed that the IL-7/IL-7R pathway played a direct inhibitory role in osteoclast differentiation via STAT5 signalling pathway in a RANKL-induced manner. We applied flow cytometry to analyse the effect of IL-7 on T-cell RANKL expression and found that IL-7/IL-7R pathway had an indirect role in the osteoclast differentiation process by enhancing the RANKL expression on T cells. In conclusion, the IL-7/IL-7R pathway exhibited a dual effect on osteoclastogenesis of CIA mice by interacting with osteoimmunology processes and could be a novel therapeutic target for autoimmune diseases such as RA.
Collapse
Affiliation(s)
- Haiyan Xu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Cai
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zijian Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Zhang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guojue Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongli Xie
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongshuai Jiang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyang Yuan
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Nie
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Wang M, Xia F, Wei Y, Wei X. Molecular mechanisms and clinical management of cancer bone metastasis. Bone Res 2020; 8:30. [PMID: 32793401 PMCID: PMC7391760 DOI: 10.1038/s41413-020-00105-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
As one of the most common metastatic sites of malignancies, bone has a unique microenvironment that allows metastatic tumor cells to grow and flourish. The fenestrated capillaries in the bone, bone matrix, and bone cells, including osteoblasts and osteoclasts, together maintain the homeostasis of the bone microenvironment. In contrast, tumor-derived factors act on bone components, leading to subsequent bone resorption or excessive bone formation. The various pathways involved also provide multiple targets for therapeutic strategies against bone metastases. In this review, we summarize the current understanding of the mechanism of bone metastases. Based on the general process of bone metastases, we specifically highlight the complex crosstalk between tumor cells and the bone microenvironment and the current management of cancer bone metastases.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| |
Collapse
|
14
|
Zehentmeier S, Pereira JP. Cell circuits and niches controlling B cell development. Immunol Rev 2020; 289:142-157. [PMID: 30977190 DOI: 10.1111/imr.12749] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 02/06/2023]
Abstract
Studies over the last decade uncovered overlapping niches for hematopoietic stem cells (HSCs), multipotent progenitor cells, common lymphoid progenitors, and early B cell progenitors. HSC and lymphoid niches are predominantly composed by mesenchymal progenitor cells (MPCs) and by a small subset of endothelial cells. Niche cells create specialized microenvironments through the concomitant production of short-range acting cell-fate determining cytokines such as interleukin (IL)-7 and stem cell factor and the potent chemoattractant C-X-C motif chemokine ligand 12. This type of cellular organization allows for the cross-talk between hematopoietic stem and progenitor cells with niche cells, such that niche cell activity can be regulated by the quality and quantity of hematopoietic progenitors being produced. For example, preleukemic B cell progenitors and preB acute lymphoblastic leukemias interact directly with MPCs, and downregulate IL-7 expression and the production of non-leukemic lymphoid cells. In this review, we discuss a novel model of B cell development that is centered on cellular circuits formed between B cell progenitors and lymphopoietic niches.
Collapse
Affiliation(s)
- Sandra Zehentmeier
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - João P Pereira
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
15
|
Novak S, Roeder E, Kalinowski J, Jastrzebski S, Aguila HL, Lee SK, Kalajzic I, Lorenzo JA. Osteoclasts Derive Predominantly from Bone Marrow-Resident CX 3CR1 + Precursor Cells in Homeostasis, whereas Circulating CX 3CR1 + Cells Contribute to Osteoclast Development during Fracture Repair. THE JOURNAL OF IMMUNOLOGY 2020; 204:868-878. [PMID: 31915261 DOI: 10.4049/jimmunol.1900665] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/12/2019] [Indexed: 01/10/2023]
Abstract
Osteoclasts (OC) originate from either bone marrow (BM)-resident or circulating myeloid OC progenitors (OCP) expressing the receptor CX3CR1. Multiple lines of evidence argue that OCP in homeostasis and inflammation differ. We investigated the relative contributions of BM-resident and circulating OCP to osteoclastogenesis during homeostasis and fracture repair. Using CX3CR1-EGFP/TRAP tdTomato mice, we found CX3CR1 expression in mononuclear cells, but not in multinucleated TRAP+ OC. However, CX3CR1-expressing cells generated TRAP+ OC on bone within 5 d in CX3CR1CreERT2/Ai14 tdTomato reporter mice. To define the role that circulating cells play in osteoclastogenesis during homeostasis, we parabiosed TRAP tdTomato mice (CD45.2) on a C57BL/6 background with wild-type (WT) mice (CD45.1). Flow cytometry (CD45.1/45.2) demonstrated abundant blood cell mixing between parabionts after 2 wk. At 4 wk, there were numerous tdTomato+ OC in the femurs of TRAP tdTomato mice but almost none in WT mice. Similarly, cultured BM stimulated to form OC demonstrated multiple fluorescent OC in cell cultures from TRAP tdTomato mice, but not from WT mice. Finally, flow cytometry confirmed low-level engraftment of BM cells between parabionts but significant engraftment in the spleens. In contrast, during fracture repair, we found that circulating CX3CR1+ cells migrated to bone, lost expression of CX3CR1, and became OC. These data demonstrate that OCP, but not mature OC, express CX3CR1 during both homeostasis and fracture repair. We conclude that, in homeostasis mature OC derive predominantly from BM-resident OCP, whereas during fracture repair, circulating CX3CR1+ cells can become OC.
Collapse
Affiliation(s)
- Sanja Novak
- Department of Reconstructive Sciences, UConn Health, Farmington, CT 06030
| | - Emilie Roeder
- Department of Reconstructive Sciences, UConn Health, Farmington, CT 06030
| | | | | | - Hector L Aguila
- Department of Immunology, UConn Health, Farmington, CT 06030
| | - Sun-Kyeong Lee
- University of Connecticut Center on Aging, UConn Health, Farmington, CT 06030; and
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, CT 06030
| | - Joseph A Lorenzo
- Department of Medicine, UConn Health, Farmington, CT 06030; .,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| |
Collapse
|
16
|
Jastrzebski S, Kalinowski J, Mun S, Shin B, Adapala NS, Jacome-Galarza CE, Mirza F, Aguila HL, Drissi H, Sanjay A, Canalis E, Lee SK, Lorenzo JA. Protease-Activated Receptor 1 Deletion Causes Enhanced Osteoclastogenesis in Response to Inflammatory Signals through a Notch2-Dependent Mechanism. THE JOURNAL OF IMMUNOLOGY 2019; 203:105-116. [PMID: 31109956 DOI: 10.4049/jimmunol.1801032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/25/2019] [Indexed: 12/29/2022]
Abstract
We found that protease-activated receptor 1 (PAR1) was transiently induced in cultured osteoclast precursor cells. Therefore, we examined the bone phenotype and response to resorptive stimuli of PAR1-deficient (knockout [KO]) mice. Bones and bone marrow-derived cells from PAR1 KO and wild-type (WT) mice were assessed using microcomputed tomography, histomorphometry, in vitro cultures, and RT-PCR. Osteoclastic responses to TNF-α (TNF) challenge in calvaria were analyzed with and without a specific neutralizing Ab to the Notch2-negative regulatory region (N2-NRR Ab). In vivo under homeostatic conditions, there were minimal differences in bone mass or bone cells between PAR1 KO and WT mice. However, PAR1 KO myeloid cells demonstrated enhanced osteoclastogenesis in response to receptor activator of NF-κB ligand (RANKL) or the combination of RANKL and TNF. Strikingly, in vivo osteoclastogenic responses of PAR1 KO mice to TNF were markedly enhanced. We found that N2-NRR Ab reduced TNF-induced osteoclastogenesis in PAR1 KO mice to WT levels without affecting WT responses. Similarly, in vitro N2-NRR Ab reduced RANKL-induced osteoclastogenesis in PAR1 KO cells to WT levels without altering WT responses. We conclude that PAR1 functions to limit Notch2 signaling in responses to RANKL and TNF and moderates osteoclastogenic response to these cytokines. This effect appears, at least in part, to be cell autonomous because enhanced osteoclastogenesis was seen in highly purified PAR1 KO osteoclast precursor cells. It is likely that this pathway is involved in regulating the response of bone to diseases associated with inflammatory signals.
Collapse
Affiliation(s)
| | | | - Sehwan Mun
- Center on Aging, UConn Health, Farmington, CT 06030
| | - Bongjin Shin
- Center on Aging, UConn Health, Farmington, CT 06030
| | | | | | - Faryal Mirza
- Department of Medicine, UConn Health, Farmington, CT 06030
| | | | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| | - Ernesto Canalis
- Department of Medicine, UConn Health, Farmington, CT 06030.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| | | | - Joseph A Lorenzo
- Department of Medicine, UConn Health, Farmington, CT 06030; .,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030
| |
Collapse
|
17
|
Green AC, Rudolph-Stringer V, Chantry AD, Wu JY, Purton LE. Mesenchymal lineage cells and their importance in B lymphocyte niches. Bone 2019; 119:42-56. [PMID: 29183783 PMCID: PMC11488667 DOI: 10.1016/j.bone.2017.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 02/06/2023]
Abstract
Early B lymphopoiesis occurs in the bone marrow and is reliant on interactions with numerous cell types in the bone marrow microenvironment, particularly those of the mesenchymal lineage. Each cellular niche that supports the distinct stages of B lymphopoiesis is unique. Different cell types and signaling molecules are important for the progressive stages of B lymphocyte differentiation. Cells expressing CXCL12 and IL-7 have long been recognized as having essential roles in facilitating progression through stages of B lymphopoiesis. Recently, a number of other factors that extrinsically mediate B lymphopoiesis (positively or negatively) have been identified. In addition, the use of transgenic mouse models to delete specific genes in mesenchymal lineage cells has further contributed to our understanding of how B lymphopoiesis is regulated in the bone marrow. This review will cover the current understanding of B lymphocyte niches in the bone marrow and key extrinsic molecules and signaling pathways involved in these niches, with a focus on how mesenchymal lineage cells regulate B lymphopoiesis.
Collapse
Affiliation(s)
- Alanna C Green
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia; Sheffield Myeloma Research Team, Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK; The Mellanby Centre for Bone Research, Sheffield, UK.
| | - Victoria Rudolph-Stringer
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Andrew D Chantry
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK; The Mellanby Centre for Bone Research, Sheffield, UK
| | - Joy Y Wu
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Louise E Purton
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia.
| |
Collapse
|
18
|
The Differentiation Balance of Bone Marrow Mesenchymal Stem Cells Is Crucial to Hematopoiesis. Stem Cells Int 2018; 2018:1540148. [PMID: 29765406 PMCID: PMC5903338 DOI: 10.1155/2018/1540148] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/21/2018] [Indexed: 01/20/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs), the important component and regulator of bone marrow microenvironment, give rise to hematopoietic-supporting stromal cells and form hematopoietic niches for hematopoietic stem cells (HSCs). However, how BMSC differentiation affects hematopoiesis is poorly understood. In this review, we focus on the role of BMSC differentiation in hematopoiesis. We discussed the role of BMSCs and their progeny in hematopoiesis. We also examine the mechanisms that cause differentiation bias of BMSCs in stress conditions including aging, irradiation, and chemotherapy. Moreover, the differentiation balance of BMSCs is crucial to hematopoiesis. We highlight the negative effects of differentiation bias of BMSCs on hematopoietic recovery after bone marrow transplantation. Keeping the differentiation balance of BMSCs is critical for hematopoietic recovery. This review summarises current understanding about how BMSC differentiation affects hematopoiesis and its potential application in improving hematopoietic recovery after bone marrow transplantation.
Collapse
|
19
|
Wang Y, Xiao M, Tao C, Chen J, Wang Z, Yang J, Chen Z, Zou Z, Liu A, Cai D, Jiang Y, Ding C, Li M, Bai X. Inactivation of mTORC1 Signaling in Osterix-Expressing Cells Impairs B-cell Differentiation. J Bone Miner Res 2018; 33:732-742. [PMID: 29206332 DOI: 10.1002/jbmr.3352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/23/2017] [Accepted: 11/26/2017] [Indexed: 01/17/2023]
Abstract
Osteoblasts provide a microenvironmental niche for B-cell commitment and maturation in the bone marrow (BM). Any abnormity of osteoblasts function may result in the defect of B lymphopoiesis. Signaling from mechanistic target of rapamycin complex 1 (mTORC1) has been implicated in regulating the expansion and differentiation of osteoblasts. Thus, we raise a hypothesis that mTORC1 signaling in osteoblasts plays a vital role in B-cell development. Inactivation of mTORC1 in osterix-expressing cells (mainly osteoblast lineage) through Osx-Cre-directed deletion of Raptor (an mTORC1-specific component) resulted in a reduction in the total B-cell population in the BM, which was due to a block in early B-cell development from the pro-B to pre-B cell stage. Further mechanistic studies revealed that this defect was the result of reduction of interleukin-7 (IL-7) expression in osterix-expressing immature osteoblasts, which caused the abnormality of IL-7/Stat5 signaling in early B lymphocytes, leading to an increased apoptosis of pre-B plus immature B cells. In vitro and in vivo studies demonstrated that the addition of exogenous IL-7 partially restored B lymphopoiesis in the BM of Raptor mutant mice. Furthermore, total BM cells cultured in conditioned media from Raptor null immature osteoblasts or media with anti-IL-7 neutralizing antibody failed to differentiate into pre-B and immature B cells, indicating that inactivation of mTORC1 in immature osteoblast cannot fully support normal B-cell development. Taken together, these findings demonstrate a novel role for mTORC1 in the regulation of bone marrow environments that support B-cell differentiation via regulating IL-7 expression. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yongkui Wang
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Min Xiao
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chen Tao
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jing Chen
- SunYat-sen University Cancer Center, Guangzhou, China
| | - Zhenyu Wang
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jun Yang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenguo Chen
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhipeng Zou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Anling Liu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Daozhang Cai
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, USA
| | - Changhai Ding
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mangmang Li
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of Osteoclast Differentiation by Cytokine Networks. Immune Netw 2018; 18:e8. [PMID: 29503739 PMCID: PMC5833125 DOI: 10.4110/in.2018.18.e8] [Citation(s) in RCA: 309] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 12/20/2022] Open
Abstract
Cytokines play a pivotal role in maintaining bone homeostasis. Osteoclasts (OCs), the sole bone resorbing cells, are regulated by numerous cytokines. Macrophage colony-stimulating factor and receptor activator of NF-κB ligand play a central role in OC differentiation, which is also termed osteoclastogenesis. Osteoclastogenic cytokines, including tumor necrosis factor-α, IL-1, IL-6, IL-7, IL-8, IL-11, IL-15, IL-17, IL-23, and IL-34, promote OC differentiation, whereas anti-osteoclastogenic cytokines, including interferon (IFN)-α, IFN-β, IFN-γ, IL-3, IL-4, IL-10, IL-12, IL-27, and IL-33, downregulate OC differentiation. Therefore, dynamic regulation of osteoclastogenic and anti-osteoclastogenic cytokines is important in maintaining the balance between bone-resorbing OCs and bone-forming osteoblasts (OBs), which eventually affects bone integrity. This review outlines the osteoclastogenic and anti-osteoclastogenic properties of cytokines with regard to osteoimmunology, and summarizes our current understanding of the roles these cytokines play in osteoclastogenesis.
Collapse
Affiliation(s)
| | - Hyeongseok Yun
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Hyunjong Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
21
|
Yeon Won H, Hwan Mun S, Shin B, Lee SK. Contradictory Role of CD97 in Basal and Tumor Necrosis Factor-Induced Osteoclastogenesis In Vivo. Arthritis Rheumatol 2017; 68:1301-13. [PMID: 26663852 DOI: 10.1002/art.39538] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 12/01/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE CD97, a member of the 7-transmembrane epidermal growth factor family of adhesion G protein-coupled receptors, is expressed on various cell types. This study was undertaken to elucidate the functions of CD97 in bone and inflammation in an experimental mouse model, by examining the effect of CD97 on osteoclastogenesis in vitro, characterizing the skeletal phenotype of CD97-deficient (CD97-knockout [KO]) mice, and assessing the responses to tumor necrosis factor (TNF) treatment. METHODS Femoral tissue and bone marrow (BM)-derived cells from CD97-KO and wild-type (WT) mice were assessed using histomorphometric analyses, in vitro cultures, and reverse transcription-polymerase chain reaction. Serum cytokine and chemokine levels in the presence or absence of TNF challenge were analyzed by multiplex assay. RESULTS In cultures of mouse BM-derived macrophages in vitro, RANKL induced the expression of CD97. In vivo, the trabecular bone volume of the femurs of female CD97-KO mice was increased, and this was associated with a decrease in the number of osteoclasts. Compared to WT mice, CD97-KO mice had a reduced potential to form osteoclast-like cells in vitro. Furthermore, TNF treatment augmented the formation of osteoclasts in the calvaria of CD97-KO mice in vivo, by increasing the production of RANKL and other cytokines and chemokines and by reducing the production of osteoprotegerin by calvarial cells. CONCLUSION These findings demonstrate that CD97 is a positive regulator of osteoclast-like cell differentiation, a mechanism that influences bone homeostasis. However, the presence of CD97 may be essential to suppress the initial osteoclastogenesis that occurs in response to acute and local inflammatory stimuli.
Collapse
Affiliation(s)
| | | | - Bongjin Shin
- University of Connecticut Health Center, Farmington
| | | |
Collapse
|
22
|
|
23
|
Laurent MR, Dubois V, Claessens F, Verschueren SMP, Vanderschueren D, Gielen E, Jardí F. Muscle-bone interactions: From experimental models to the clinic? A critical update. Mol Cell Endocrinol 2016; 432:14-36. [PMID: 26506009 DOI: 10.1016/j.mce.2015.10.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
Bone is a biomechanical tissue shaped by forces from muscles and gravitation. Simultaneous bone and muscle decay and dysfunction (osteosarcopenia or sarco-osteoporosis) is seen in ageing, numerous clinical situations including after stroke or paralysis, in neuromuscular dystrophies, glucocorticoid excess, or in association with vitamin D, growth hormone/insulin like growth factor or sex steroid deficiency, as well as in spaceflight. Physical exercise may be beneficial in these situations, but further work is still needed to translate acceptable and effective biomechanical interventions like vibration therapy from animal models to humans. Novel antiresorptive and anabolic therapies are emerging for osteoporosis as well as drugs for sarcopenia, cancer cachexia or muscle wasting disorders, including antibodies against myostatin or activin receptor type IIA and IIB (e.g. bimagrumab). Ideally, increasing muscle mass would increase muscle strength and restore bone loss from disuse. However, the classical view that muscle is unidirectionally dominant over bone via mechanical loading is overly simplistic. Indeed, recent studies indicate a role for neuronal regulation of not only muscle but also bone metabolism, bone signaling pathways like receptor activator of nuclear factor kappa-B ligand (RANKL) implicated in muscle biology, myokines affecting bone and possible bone-to-muscle communication. Moreover, pharmacological strategies inducing isolated myocyte hypertrophy may not translate into increased muscle power because tendons, connective tissue, neurons and energy metabolism need to adapt as well. We aim here to critically review key musculoskeletal molecular pathways involved in mechanoregulation and their effect on the bone-muscle unit as a whole, as well as preclinical and emerging clinical evidence regarding the effects of sarcopenia therapies on osteoporosis and vice versa.
Collapse
Affiliation(s)
- Michaël R Laurent
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dubois
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sabine M P Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Rehabilitation Science, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Evelien Gielen
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
24
|
Abstract
Osteoporosis develops when the rate of osteoclastic bone breakdown (resorption) exceeds that of osteoblastic bone formation, which leads to loss of BMD and deterioration of bone structure and strength. Osteoporosis increases the risk of fragility fractures, a cause of substantial morbidity and mortality, especially in elderly patients. This imbalance between bone formation and bone resorption is brought about by natural ageing processes, but is frequently exacerbated by a number of pathological conditions. Of importance to the aetiology of osteoporosis are findings over the past two decades attesting to a deep integration of the skeletal system with the immune system (the immuno-skeletal interface (ISI)). Although protective of the skeleton under physiological conditions, the ISI might contribute to bone destruction in a growing number of pathophysiological states. Although numerous research groups have investigated how the immune system affects basal and pathological osteoclastic bone resorption, recent findings suggest that the reach of the adaptive immune response extends to the regulation of osteoblastic bone formation. This Review examines the evolution of the field of osteoimmunology and how advances in our understanding of the ISI might lead to novel approaches to prevent and treat bone loss, and avert fractures.
Collapse
Affiliation(s)
- M Neale Weitzmann
- The Atlanta Department of Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, Georgia, 30033, USA
- Department of Medicine, Division of Endocrinology and Metabolism and Lipids, Emory University School of Medicine, 101 Woodruff Circle, 1305 WMB, Atlanta, Georgia 30322, USA
| | - Ighovwerha Ofotokun
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, 49 Jesse Hill Jr Drive, Atlanta, Georgia 30303, USA
- Grady Healthcare System, 80 Jesse Hill Jr Drive SE, Atlanta, Georgia, 30303, USA
| |
Collapse
|
25
|
Paglia DN, Yang X, Kalinowski J, Jastrzebski S, Drissi H, Lorenzo J. Runx1 Regulates Myeloid Precursor Differentiation Into Osteoclasts Without Affecting Differentiation Into Antigen Presenting or Phagocytic Cells in Both Males and Females. Endocrinology 2016; 157:3058-69. [PMID: 27267711 PMCID: PMC4967120 DOI: 10.1210/en.2015-2037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Runt-related transcription factor 1 (Runx1), a master regulator of hematopoiesis, is expressed in preosteoclasts. Previously we evaluated the bone phenotype of CD11b-Cre Runx1(fl/fl) mice and demonstrated enhanced osteoclasts and decreased bone mass in males. However, an assessment of the effects of Runx1 deletion in female osteoclast precursors was impossible with this model. Moreover, the role of Runx1 in myeloid cell differentiation into other lineages is unknown. Therefore, we generated LysM-Cre Runx1(fl/fl) mice, which delete Runx1 equally (∼80% deletion) in myeloid precursor cells from both sexes and examined the capacity of these cells to differentiate into osteoclasts and phagocytic and antigen-presenting cells. Both female and male LysM-Cre Runx1(fl/fl) mice had decreased trabecular bone mass (72% decrease in bone volume fraction) and increased osteoclast number (2-3 times) (P < .05) without alteration of osteoblast histomorphometric indices. We also demonstrated that loss of Runx1 in pluripotential myeloid precursors with LysM-Cre did not alter the number of myeloid precursor cells in bone marrow or their ability to differentiate into phagocytizing or antigen-presenting cells. This study demonstrates that abrogation of Runx1 in multipotential myeloid precursor cells significantly and specifically enhanced the ability of receptor activator of nuclear factor-κB ligand to stimulate osteoclast formation and fusion in female and male mice without affecting other myeloid cell fates. In turn, increased osteoclast activity in LysM-Cre Runx1(fl/fl) mice likely contributed to a decrease in bone mass. These dramatic effects were not due to increased osteoclast precursors in the deleted mutants and argue that inhibition of Runx1 in multipotential myeloid precursor cells is important for osteoclast formation and function.
Collapse
Affiliation(s)
- David N Paglia
- Departments of Orthopaedic Surgery (D.N.P., X.Y., H.D., J.L.), Medicine (J.K., S.J., J.L.), and Genetics and Genome Sciences (H.D.), University of Connecticut Health, Farmington, Connecticut 06030
| | - Xiaochuan Yang
- Departments of Orthopaedic Surgery (D.N.P., X.Y., H.D., J.L.), Medicine (J.K., S.J., J.L.), and Genetics and Genome Sciences (H.D.), University of Connecticut Health, Farmington, Connecticut 06030
| | - Judith Kalinowski
- Departments of Orthopaedic Surgery (D.N.P., X.Y., H.D., J.L.), Medicine (J.K., S.J., J.L.), and Genetics and Genome Sciences (H.D.), University of Connecticut Health, Farmington, Connecticut 06030
| | - Sandra Jastrzebski
- Departments of Orthopaedic Surgery (D.N.P., X.Y., H.D., J.L.), Medicine (J.K., S.J., J.L.), and Genetics and Genome Sciences (H.D.), University of Connecticut Health, Farmington, Connecticut 06030
| | - Hicham Drissi
- Departments of Orthopaedic Surgery (D.N.P., X.Y., H.D., J.L.), Medicine (J.K., S.J., J.L.), and Genetics and Genome Sciences (H.D.), University of Connecticut Health, Farmington, Connecticut 06030
| | - Joseph Lorenzo
- Departments of Orthopaedic Surgery (D.N.P., X.Y., H.D., J.L.), Medicine (J.K., S.J., J.L.), and Genetics and Genome Sciences (H.D.), University of Connecticut Health, Farmington, Connecticut 06030
| |
Collapse
|
26
|
Effects of myokines on bone. BONEKEY REPORTS 2016; 5:826. [PMID: 27579164 DOI: 10.1038/bonekey.2016.48] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 05/01/2016] [Indexed: 12/22/2022]
Abstract
The links between muscle and bone have been recently examined because of the increasing number of patients with osteoporosis and sarcopenia. Myokines are skeletal muscle-derived humoral cytokines and growth factors, which exert physiological and pathological functions in various distant organs, including the regulation of glucose, energy and bone metabolism. Myostatin is a crucial myokine, the expression of which is mainly limited to muscle tissues. The inhibition of myostatin signaling increases bone remodeling, bone mass and muscle mass, and it may provide a target for the treatment of both sarcopenia and osteoporosis. As myostatin is involved in osteoclast formation and bone destruction in rheumatoid arthritis, myostatin may be a target myokine for the treatment of accelerated bone resorption and joint destruction in rheumatoid arthritis. Numerous other myokines, including transforming growth factor-β, follistatin, insulin-like growth factor-I, fibroblast growth factor-2, osteoglycin, FAM5C, irisin, interleukin (IL)-6, leukemia inhibitory factor, IL-7, IL-15, monocyte chemoattractant protein-1, ciliary neurotrophic factor, osteonectin and matrix metalloproteinase 2, also affect bone cells in various manners. However, the effects of myokines on bone metabolism are largely unknown. Further research is expected to clarify the interaction between muscle and bone, which may lead to greater diagnosis and the development of the treatment for muscle and bone disorders, such as osteoporosis and sarcopenia.
Collapse
|
27
|
Bloom AC, Collins FL, Van't Hof RJ, Ryan ES, Jones E, Hughes TR, Morgan BP, Erlandsson M, Bokarewa M, Aeschlimann D, Evans BAJ, Williams AS. Deletion of the membrane complement inhibitor CD59a drives age and gender-dependent alterations to bone phenotype in mice. Bone 2016; 84:253-261. [PMID: 26721735 PMCID: PMC4764651 DOI: 10.1016/j.bone.2015.12.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/11/2015] [Accepted: 12/21/2015] [Indexed: 11/28/2022]
Abstract
Degenerative joint diseases such as osteoarthritis are characterised by aberrant region-specific bone formation and abnormal bone mineral content. A recent study suggested a role for the complement membrane attack complex in experimental models of osteoarthritis. Since CD59a is the principal regulator of the membrane attack complex in mice, we evaluated the impact of CD59a gene deletion upon maintenance of bone architecture. In vivo bone morphology analysis revealed that male CD59a-deficient mice have increased femur length and cortical bone volume, albeit with reduced bone mineral density. However, this phenomenon was not observed in female mice. Histomorphometric analysis of the trabecular bone showed increased rates of bone homeostasis, with both increased bone resorption and mineral apposition rate in CD59a-deficient male mice. When bone cells were studied in isolation, in vitro osteoclastogenesis was significantly increased in male CD59a-deficient mice, although osteoblast formation was not altered. Our data reveal, for the first time, that CD59a is a regulator of bone growth and homeostasis. CD59a ablation in male mice results in longer and wider bones, but with less density, which is likely a major contributing factor for their susceptibility to osteoarthritis. These findings increase our understanding of the role of complement regulation in degenerative arthritis.
Collapse
Affiliation(s)
- Anja C Bloom
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Fraser L Collins
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Rob J Van't Hof
- Bone Research Group, Institute of Ageing & Chronic Disease, University ofLiverpool, Liverpool, UK
| | - Elizabeth S Ryan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Emma Jones
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Timothy R Hughes
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - B Paul Morgan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Malin Erlandsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, University of Göteborg, Gothenburg, Sweden
| | - Maria Bokarewa
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, University of Göteborg, Gothenburg, Sweden
| | - Daniel Aeschlimann
- Matrix Biology and Tissue Repair, Dental School, Cardiff University, Cardiff, UK; Arthritis Research UK Centre for Biomechanics and Bioengineering, Cardiff University, Cardiff, UK
| | - Bronwen A J Evans
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, UK; Arthritis Research UK Centre for Biomechanics and Bioengineering, Cardiff University, Cardiff, UK
| | - Anwen S Williams
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK; Arthritis Research UK Centre for Biomechanics and Bioengineering, Cardiff University, Cardiff, UK.
| |
Collapse
|
28
|
Canalis E, Schilling L, Yee SP, Lee SK, Zanotti S. Hajdu Cheney Mouse Mutants Exhibit Osteopenia, Increased Osteoclastogenesis, and Bone Resorption. J Biol Chem 2015; 291:1538-1551. [PMID: 26627824 DOI: 10.1074/jbc.m115.685453] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Indexed: 11/06/2022] Open
Abstract
Notch receptors are determinants of cell fate and function and play a central role in skeletal development and bone remodeling. Hajdu Cheney syndrome, a disease characterized by osteoporosis and fractures, is associated with NOTCH2 mutations resulting in a truncated stable protein and gain-of-function. We created a mouse model reproducing the Hajdu Cheney syndrome by introducing a 6955C→T mutation in the Notch2 locus leading to a Q2319X change at the amino acid level. Notch2(Q2319X) heterozygous mutants were smaller and had shorter femurs than controls; and at 1 month of age they exhibited cancellous and cortical bone osteopenia. As the mice matured, cancellous bone volume was restored partially in male but not female mice, whereas cortical osteopenia persisted in both sexes. Cancellous bone histomorphometry revealed an increased number of osteoclasts and bone resorption, without a decrease in osteoblast number or bone formation. Osteoblast differentiation and function were not affected in Notch2(Q2319X) cells. The pre-osteoclast cell pool, osteoclast differentiation, and bone resorption in response to receptor activator of nuclear factor κB ligand in vitro were increased in Notch2(Q2319X) mutants. These effects were suppressed by the γ-secretase inhibitor LY450139. In conclusion, Notch2(Q2319X) mice exhibit cancellous and cortical bone osteopenia, enhanced osteoclastogenesis, and increased bone resorption.
Collapse
Affiliation(s)
| | | | - Siu-Pok Yee
- Cell Biology, Genetics, and; Genome Sciences Biology
| | - Sun-Kyeong Lee
- Medicine,; Center on Aging, University of Connecticut Health Center, Farmington, Connecticut 06030
| | | |
Collapse
|
29
|
Jung S, Yang HY, Lee TH. Differential expression of immunologic proteins in gingiva after socket preservation in mini pigs. J Appl Oral Sci 2015; 23:187-95. [PMID: 26018311 PMCID: PMC4428464 DOI: 10.1590/1678-775720140311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/27/2015] [Indexed: 02/03/2023] Open
Abstract
During healing following tooth extraction, inflammation and the immune response within the extraction socket are related to bone resorption.
Collapse
Affiliation(s)
- Seunggon Jung
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Hee-Young Yang
- Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Tae-Hoon Lee
- Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
30
|
Abstract
Obesity markedly increases susceptibility to a range of diseases and simultaneously undermines the viability and fate selection of haematopoietic stem cells (HSCs), and thus the kinetics of leukocyte production that is critical to innate and adaptive immunity. Considering that blood cell production and the differentiation of HSCs and their progeny is orchestrated, in part, by complex interacting signals emanating from the bone marrow microenvironment, it is not surprising that conditions that disturb bone marrow structure inevitably disrupt both the numbers and lineage-fates of these key blood cell progenitors. In addition to the increased adipose burden in visceral and subcutaneous compartments, obesity causes a marked increase in the size and number of adipocytes encroaching into the bone marrow space, almost certainly disturbing HSC interactions with neighbouring cells, which include osteoblasts, osteoclasts, mesenchymal cells and endothelial cells. As the global obesity pandemic grows, the short-term and long-term consequences of increased bone marrow adiposity on HSC lineage selection and immune function remain uncertain. This Review discusses the differentiation and function of haematopoietic cell populations, the principal physicochemical components of the bone marrow niche, and how this environment influences HSCs and haematopoiesis in general. The effect of adipocytes and adiposity on HSC and progenitor cell populations is also discussed, with the goal of understanding how obesity might compromise the core haematopoietic system.
Collapse
Affiliation(s)
- Benjamin J Adler
- Department of Biomedical Engineering, Bioengineering Building, Stony Brook University, Stony Brook, NY 11794-5281, USA
| | - Kenneth Kaushansky
- Department of Medicine, Health Sciences Centre, Stony Brook University, Stony Brook, NY 11794-8430, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Bioengineering Building, Stony Brook University, Stony Brook, NY 11794-5281, USA
| |
Collapse
|
31
|
Jiang C, Li Z, Quan H, Xiao L, Zhao J, Jiang C, Wang Y, Liu J, Gou Y, An S, Huang Y, Yu W, Zhang Y, He W, Yi Y, Chen Y, Wang J. Osteoimmunology in orthodontic tooth movement. Oral Dis 2014; 21:694-704. [PMID: 25040955 DOI: 10.1111/odi.12273] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/18/2014] [Accepted: 06/26/2014] [Indexed: 02/05/2023]
Abstract
The skeletal and immune systems share a multitude of regulatory molecules, including cytokines, receptors, signaling molecules, and signaling transducers, thereby mutually influencing each other. In recent years, several novel insights have been attained that have enhanced our current understanding of the detailed mechanisms of osteoimmunology. In orthodontic tooth movement, immune responses mediated by periodontal tissue under mechanical force induce the generation of inflammatory responses with consequent alveolar bone resorption, and many regulators are involved in this process. In this review, we take a closer look at the cellular/molecular mechanisms and signaling involved in osteoimmunology and at relevant research progress in the context of the field of orthodontic tooth movement.
Collapse
Affiliation(s)
- C Jiang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Z Li
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - H Quan
- Qingdao First Sanatorium of Jinan Military Distract of PLA, Qingdao, Shandong, China
| | - L Xiao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Zhao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - C Jiang
- Department of Prosthodontics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Y Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Liu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Y Gou
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S An
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Huang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - W Yu
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Zhang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - W He
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Yi
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Chen
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Wang
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, Sichuan, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Adler BJ, Green DE, Pagnotti GM, Chan ME, Rubin CT. High fat diet rapidly suppresses B lymphopoiesis by disrupting the supportive capacity of the bone marrow niche. PLoS One 2014; 9:e90639. [PMID: 24595332 PMCID: PMC3942453 DOI: 10.1371/journal.pone.0090639] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 02/03/2014] [Indexed: 12/15/2022] Open
Abstract
The bone marrow (BM) niche is the primary site of hematopoiesis, and cues from this microenvironment are critical to maintain hematopoiesis. Obesity increases lifetime susceptibility to a host of chronic diseases, and has been linked to defective leukogenesis. The pressures obesity exerts on hematopoietic tissues led us to study the effects of a high fat diet (HFD: 60% Kcal from fat) on B cell development in BM. Seven week old male C57Bl/6J mice were fed either a high fat (HFD) or regular chow (RD) diet for periods of 2 days, 1 week and 6 weeks. B-cell populations (B220+) were not altered after 2 d of HFD, within 1 w B-cell proportions were reduced by −10%, and by 6 w by −25% as compared to RD (p<0.05). BM RNA was extracted to track the expression of B-cell development markers Il-7, Ebf-1 and Pax-5. At 2 d, the expression of Il-7 and Ebf-1 were reduced by −20% (p = 0.08) and −11% (p = 0.06) whereas Pax-5 was not significantly impacted. At one week, however, the expressions of Il-7, Ebf-1, and Pax-5 in HFD mice fell by -19%, −20% and −16%, and by six weeks were further reduced to −23%, −29% and −34% as compared to RD (p<0.05 for all), a suppression paralleled by a +363% increase in adipose encroachment within the marrow space (p<0.01). Il-7 is a critical factor in the early B-cell lineage which is secreted by supportive cells in the BM niche, and is necessary for B-cell commitment. These data indicate that BM Il-7 expression, and by extension B-cell differentiation, are rapidly impaired by HFD. The trend towards suppressed expression of Il-7 following only 2 d of HFD demonstrates how susceptible the BM niche, and the cells which rely on it, are to diet, which ultimately could contribute to disease susceptibility in metabolic disorders such as obesity.
Collapse
Affiliation(s)
- Benjamin J. Adler
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Danielle E. Green
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Gabriel M. Pagnotti
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - M. Ete Chan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Clinton T. Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
33
|
Panaroni C, Tzeng YS, Saeed H, Wu JY. Mesenchymal progenitors and the osteoblast lineage in bone marrow hematopoietic niches. Curr Osteoporos Rep 2014; 12:22-32. [PMID: 24477415 PMCID: PMC4077781 DOI: 10.1007/s11914-014-0190-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The bone marrow cavity is essential for the proper development of the hematopoietic system. In the last few decades, it has become clear that mesenchymal stem/progenitor cells as well as cells of the osteoblast lineage, besides maintaining bone homeostasis, are also fundamental regulators of bone marrow hematopoiesis. Several studies have demonstrated the direct involvement of mesenchymal and osteoblast lineage cells in the maintenance and regulation of supportive microenvironments necessary for quiescence, self-renewal and differentiation of hematopoietic stem cells. In addition, specific niches have also been identified within the bone marrow for maturing hematopoietic cells. Here we will review recent findings that have highlighted the roles of mesenchymal progenitors and cells of the osteoblast lineage in regulating distinct stages of hematopoiesis.
Collapse
Affiliation(s)
- Cristina Panaroni
- Division of Endocrinology, Stanford University School of Medicine, 300 Pasteur Dr., S-025, Stanford, CA, 94305, USA
| | | | | | | |
Collapse
|
34
|
Soung DY, Kalinowski J, Baniwal SK, Jacome-Galarza CE, Frenkel B, Lorenzo J, Drissi H. Runx1-mediated regulation of osteoclast differentiation and function. Mol Endocrinol 2014; 28:546-53. [PMID: 24606124 DOI: 10.1210/me.2013-1305] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Excessive bone resorption is the cause of several metabolic bone diseases including osteoporosis. Thus, identifying factors that can inhibit osteoclast formation and/or activity may define new drug targets that can be used to develop novel therapies for these conditions. Emerging evidence demonstrates that the master regulator of hematopoiesis, Runx1, is expressed in preosteoclasts and may influence skeletal health. To examine the potential role of Runx1 in osteoclast formation and function, we deleted its expression in myeloid osteoclast precursors by crossing Runx1 floxed mice (Runx1(F/F)) with CD11b-Cre transgenic mice. Mice lacking Runx1 in preosteoclasts (CD11b-Cre;Runx1(F/F)) exhibited significant loss of femoral trabecular and cortical bone mass compared with that in Cre-negative mice. In addition, serum levels of collagen type 1 cross-linked C-telopeptide, a biomarker of osteoclast-mediated bone resorption, were significantly elevated in CD11b-Cre;Runx1(F/F) mice compared with those in Runx1(F/F) mice. Tartrate-resistant acid phosphatase-positive osteoclasts that differentiated from bone marrow cells of CD11b-Cre;Runx1(F/F) mice in vitro were larger, were found in greater numbers, and had increased bone resorbing activity than similarly cultured cells from Runx1(F/F) mice. CD11b-Cre;Runx1(F/F) bone marrow cells that were differentiated into osteoclasts in vitro also had elevated mRNA levels of osteoclast-related genes including vacuolar ATPase D2, cathepsin K, matrix metalloproteinase 9, calcitonin receptor, osteoclast-associated receptor, nuclear factor of activated T cells cytoplasmic 1, and cFos. These data indicate that Runx1 expression in preosteoclasts negatively regulates osteoclast formation and activity and contributes to overall bone mass.
Collapse
Affiliation(s)
- Do Y Soung
- New England Musculoskeletal Institute (D.Y.S., J.K., C.E.J.-G., J.L., H.D.), Department of Orthopaedic Surgery (H.D.), and Department of Medicine (C.E.J.-G.), University of Connecticut Health Center, Farmington, Connecticut 06030; and Department of Orthopaedic Surgery and Biochemistry and Molecular Biology (S.K.B., B.F.), University of Southern California, Los Angeles, California 90089
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Panaroni C, Wu JY. Interactions between B lymphocytes and the osteoblast lineage in bone marrow. Calcif Tissue Int 2013; 93:261-8. [PMID: 23839529 PMCID: PMC3762579 DOI: 10.1007/s00223-013-9753-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 05/22/2013] [Indexed: 10/26/2022]
Abstract
The regulatory effects of the immune system on the skeleton during homeostasis and activation have been appreciated for years. In the past decade it has become evident that bone tissue can also regulate immune cell development. In the bone marrow, the differentiation of hematopoietic progenitors requires specific microenvironments, called "niches," provided by various subsets of stromal cells, many of which are of mesenchymal origin. Among these stromal cell populations, cells of the osteoblast lineage serve a supportive function in the maintenance of normal hematopoiesis, and B lymphopoiesis in particular. Within the osteoblast lineage, distinct differentiation stages exert differential regulatory effects on hematopoietic development. In this review we will highlight the critical role of osteoblast progenitors in the perivascular B lymphocyte niche.
Collapse
Affiliation(s)
- Cristina Panaroni
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, CA 94305, USA
| | | |
Collapse
|
37
|
Sweeney E, Roberts D, Lin A, Guldberg R, Jacenko O. Defective endochondral ossification-derived matrix and bone cells alter the lymphopoietic niche in collagen X mouse models. Stem Cells Dev 2013; 22:2581-95. [PMID: 23656481 DOI: 10.1089/scd.2012.0387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Despite the appreciated interdependence of skeletal and hematopoietic development, the cell and matrix components of the hematopoietic niche remain to be fully defined. Utilizing mice with disrupted function of collagen X (ColX), a major hypertrophic cartilage matrix protein associated with endochondral ossification, our data identified a cytokine defect in trabecular bone cells at the chondro-osseous hematopoietic niche as a cause for aberrant B lymphopoiesis in these mice. Specifically, analysis of ColX transgenic and null mouse chondro-osseous regions via micro-computed tomography revealed an altered trabecular bone environment. Additionally, cocultures with hematopoietic and chondro-osseous cell types highlighted impaired hematopoietic support by ColX transgenic and null mouse derived trabecular bone cells. Further, cytokine arrays with conditioned media from the trabecular osteoblast cocultures suggested an aberrant hematopoietic cytokine milieu within the chondro-osseous niche of the ColX deficient mice. Accordingly, B lymphopoiesis was rescued in the ColX mouse derived trabecular osteoblast cocultures with interlukin-7, stem cell factor, and stromal derived factor-1 supplementation. Moreover, B cell development was restored in vivo after injections of interlukin-7. These data support our hypothesis that endrochondrally-derived trabecular bone cells and matrix constituents provide cytokine-rich niches for hematopoiesis. Furthermore, this study contributes to the emerging concept that niche defects may underlie certain immuno-osseous and hematopoietic disorders.
Collapse
Affiliation(s)
- Elizabeth Sweeney
- 1 Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | | | | | | |
Collapse
|
38
|
Mun SH, Won HY, Hernandez P, Aguila HL, Lee SK. Deletion of CD74, a putative MIF receptor, in mice enhances osteoclastogenesis and decreases bone mass. J Bone Miner Res 2013; 28:948-59. [PMID: 23044992 PMCID: PMC3563845 DOI: 10.1002/jbmr.1787] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/26/2012] [Accepted: 10/01/2012] [Indexed: 12/24/2022]
Abstract
CD74 is a type II transmembrane protein that can act as a receptor for macrophage migration inhibitory factor (MIF) and plays a role in MIF-regulated responses. We reported that MIF inhibited osteoclast formation and MIF knockout (KO) mice had decreased bone mass. We therefore examined if CD74 was involved in the ability of MIF to alter osteoclastogenesis in cultured bone marrow (BM) from wild-type (WT) and CD74-deficient (KO) male mice. We also measured the bone phenotype of CD74 KO male mice. Bone mass in the femur of 8-week-old mice was measured by micro-computed tomography and histomorphometry. Bone marrow cells from CD74 KO mice formed 15% more osteoclast-like cells (OCLs) with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL) (both at 30 ng/mL) compared to WT. Addition of MIF to WT cultures inhibited OCL formation by 16% but had no effect on CD74KO cultures. The number of colony forming unit granulocyte-macrophage (CFU-GM) in the bone marrow of CD74 KO mice was 26% greater than in WT controls. Trabecular bone volume (TBV) in the femurs of CD74 KO male mice was decreased by 26% compared to WT. In addition, cortical area and thickness were decreased by 14% and 11%, respectively. Histomorphometric analysis demonstrated that tartrate-resistant acid phosphatase (TRAP)(+) osteoclast number and area were significantly increased in CD74 KO by 35% and 43%, respectively compared to WT. Finally, we examined the effect of MIF on RANKL-induced-signaling pathways in bone marrow macrophage (BMM) cultures. MIF treatment decreased RANKL-induced nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) and c-Fos protein in BMM cultures by 70% and 41%, respectively. Our data demonstrate that CD74 is required for MIF to affect in vitro osteoclastogenesis. Further, the bone phenotype of CD74 KO mice is similar to that of MIF KO mice. MIF treatment of WT cultures suppressed RANKL-induced activator protein 1 (AP-1) expression, which resulted in decreased osteoclast differentiation in vitro. We propose that CD74 plays a critical role in the MIF inhibition of osteoclastogenesis.
Collapse
Affiliation(s)
- Se Hwan Mun
- UCONN Center on Aging, University of Connecticut Health Center, Farmington, CT 06030-1835, USA
| | | | | | | | | |
Collapse
|
39
|
Zupan J, Jeras M, Marc J. Osteoimmunology and the influence of pro-inflammatory cytokines on osteoclasts. Biochem Med (Zagreb) 2013; 23:43-63. [PMID: 23457765 PMCID: PMC3900089 DOI: 10.11613/bm.2013.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bone and immune system are functionally interconnected. Immune and bone cells derive from same progenitors in the bone marrow, they share a common microenvironment and are being influenced by similar mediators. The evidence on increased bone resorption associated with inappropriate activation of T cells such as during inflammation, is well established. However, the molecular mechanisms beyond this clinical observation have begun to be intensively studied with the advancement of osteoimmunology. Now days, we have firm evidence on the influence of numerous proinflammatory cytokines on bone cells, with the majority of data focused on osteoclasts, the bone resorbing cells. It has been shown that some proinflammatory cytokines could possess osteoclastogenic and/or anti-osteoclastogenic properties and can target osteoclasts directly or via receptor activator of nuclear factor κB (RANK)/RANK ligand(RANKL)/osteoprotegerin (OPG) system. Several studies have reported opposing data regarding (anti)osteoclastogenic properties of these cytokines. Therefore, the first part of this review is summarizing current evidence on the influence of pro-inflammatory cytokines on osteoclasts and thus on bone resorption. In the second part, the evidence on the role of pro-inflammatory cytokines in osteoporosis and osteoarthritis is reviewed to show that unravelling the mechanisms beyond such complex bone diseases, is almost impossible without considering skeletal and immune systems as an indivisible integrated system.
Collapse
Affiliation(s)
- Janja Zupan
- University of Ljubljana, Faculty of Pharmacy, Department of Clinical Biochemistry, Ljubljana, Slovenia
| | | | | |
Collapse
|
40
|
Weitzmann MN. The Role of Inflammatory Cytokines, the RANKL/OPG Axis, and the Immunoskeletal Interface in Physiological Bone Turnover and Osteoporosis. SCIENTIFICA 2013; 2013:125705. [PMID: 24278766 PMCID: PMC3820310 DOI: 10.1155/2013/125705] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 12/24/2012] [Indexed: 05/30/2023]
Abstract
Although it has long been recognized that inflammation, a consequence of immune-driven processes, significantly impacts bone turnover, the degree of centralization of skeletal and immune functions has begun to be dissected only recently. It is now recognized that formation of osteoclasts, the bone resorbing cells of the body, is centered on the key osteoclastogenic cytokine, receptor activator of NF- κ B ligand (RANKL). Although numerous inflammatory cytokines are now recognized to promote osteoclast formation and skeletal degradation, with just a few exceptions, RANKL is now considered to be the final downstream effector cytokine that drives osteoclastogenesis and regulates osteoclastic bone resorption. The biological activity of RANKL is moderated by its physiological decoy receptor, osteoprotegerin (OPG). New discoveries concerning the sources and regulation of RANKL and OPG in physiological bone turnover as well as under pathological (osteoporotic) conditions continue to be made, opening a window to the complex regulatory processes that control skeletal integrity and the depth of integration of the skeleton within the immune response. This paper will examine the interconnection between bone turnover and the immune system and the implications thereof for physiological and pathological bone turnover.
Collapse
Affiliation(s)
- M. Neale Weitzmann
- Atlanta Department of Veterans Affairs Medical Center, Decatur, GA 30033, USA
- Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, 101 Woodruff Circle, 1305 WMRB, Atlanta, GA 30322, USA
| |
Collapse
|
41
|
Horowitz MC, Fretz JA, Lorenzo JA. How B cells influence bone biology in health and disease. Bone 2010; 47:472-9. [PMID: 20601290 PMCID: PMC2941392 DOI: 10.1016/j.bone.2010.06.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/13/2010] [Accepted: 06/14/2010] [Indexed: 12/27/2022]
Abstract
It is now well established that important regulatory interactions occur between the cells in the hematopoietic, immune and skeletal systems (osteoimmunology). B lymphocytes (B cells) are responsible for the generation and production of antibodies or immunoglobulins in the body. Together with T cells these lymphocytes comprise the adaptive immune system, which allows an individual to develop specific responses to an infection and retain memory of that infection, allowing for a faster and more robust response if that same infection occurs again. In addition to this immune function, B cells have a close and multifaceted relationship with bone cells. B cells differentiate from hematopoietic stem cells (HSCs) in supportive niches found on endosteal bone surfaces. Cells in the osteoblast lineage support HSC and B cell differentiation in these niches. B cell differentiation is regulated, at least in part, by a series of transcription factors that function in a temporal manner. While these transcription factors are required for B cell differentiation, their loss causes profound changes in the bone phenotype. This is due, in part, to the close relationship between macrophage/osteoclast and B cell differentiation. Cross talk between B cells and bone cells is reciprocal with defects in the RANKL-RANK, OPG signaling axis resulting in altered bone phenotypes. While the role of B cells during normal bone remodeling appears minimal, activated B cells play an important role in many inflammatory diseases with associated bony changes. This review examines the relationship between B cells and bone cells and how that relationship affects the skeleton and hematopoiesis during health and disease.
Collapse
Affiliation(s)
- Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510, USA.
| | | | | |
Collapse
|
42
|
Abstract
It has become clear that complex interactions underlie the relationship between the skeletal and immune systems. This is particularly true for the development of immune cells in the bone marrow as well as the functions of bone cells in skeletal homeostasis and pathologies. Because these two disciplines developed independently, investigators with an interest in either often do not fully appreciate the influence of the other system on the functions of the tissue that they are studying. With these issues in mind, this review will focus on several key areas that are mediated by crosstalk between the bone and immune systems. A more complete appreciation of the interactions between immune and bone cells should lead to better therapeutic strategies for diseases that affect either or both systems.
Collapse
Affiliation(s)
- Seoung-Hoon Lee
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, U.S.A
| | - Tae-Soo Kim
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, U.S.A
| | - Yongwon Choi
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, U.S.A
| | - Joseph Lorenzo
- The Department of Medicine and the Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, U.S.A
| |
Collapse
|