1
|
Geng Z, Tong Y, Chen Y, Wang J, Liu Z, Miao J, Li R. Investigating the causal relationship between immune factors and ankylosing spondylitis: insights from a Mendelian Randomization study. Adv Rheumatol 2024; 64:89. [PMID: 39696529 DOI: 10.1186/s42358-024-00428-1] [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: 12/13/2023] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Despite previous studies indicating a close relationship between immune system and ankylosing spondylitis (AS), the causal relationship between them remains unclear. METHODS Genome-wide association data were utilized to explore the causal link between 731 immune cells and AS using a bidirectional two-sample MR approach. The data included immune cell data from Orrù et al.'s study and AS data from the FinnGen consortium. Cochran's Q test and leave-one-out checked instrument variable (IV) heterogeneity. IVW was the primary method for causal analysis, with MR-Egger and MR-PRESSO addressing horizontal pleiotropy. FDR correction was applied to both analysis directions to rectify multiple testing errors. RESULTS In our study, 22 immune phenotypes out of 731 were casually linked to AS. After excluding 5 less robust features, 17 immune factors remained, with 4 being protective and the rest posing risks. Through FDR correction, we found a significant causal relationship between HLA DR on CD14- CD16+ monocyte and AS (OR (95%CI) = 0.70(0.60 ~ 0.83), P = 2.06*10-5). In the reverse analysis with AS as exposure, potential effects on 34 immune features were discovered. After correction, we confirmed significant causal relationships between AS and two immune features, namely CD20- B cell %lymphocyte (OR (95%CI) = 1.16(1.08-1.25), P = 1.91*10-5) and CD20- B cell %B cell (OR (95%CI) = 1.17(1.09-1.26), P = 1.50*10-5). CONCLUSIONS Our study identified various features associated with AS in different types of immune cells. These findings provide important clues and a theoretical basis for further understanding the pathogenesis of AS, guiding clinical treatment, and drug design.
Collapse
Affiliation(s)
- Ziming Geng
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100141, China
| | - Yang Tong
- School of Finance, Nankai University, Tianjin, 300350, China.
| | - Yang Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Rd, Hexi District, Tianjin, 300211, China
| | - Jian Wang
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Rd, Hexi District, Tianjin, 300211, China
| | - Ziwen Liu
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Rd, Hexi District, Tianjin, 300211, China
| | - Jun Miao
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Rd, Hexi District, Tianjin, 300211, China.
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, No. 406 Jiefang South Rd, Hexi District, Tianjin, 300211, China.
| | - Ruihua Li
- Tianjin Hospital, Tianjin University, No. 406 Jiefang South Rd, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
2
|
Zhang X, Wang Y, Zheng M, Wei Q, Zhang R, Zhu K, Zhai Q, Xu Y. IMPC-based screening revealed that ROBO1 can regulate osteoporosis by inhibiting osteogenic differentiation. Front Cell Dev Biol 2024; 12:1450215. [PMID: 39439909 PMCID: PMC11494888 DOI: 10.3389/fcell.2024.1450215] [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: 07/10/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction The utilization of denosumab in treating osteoporosis highlights promising prospects for osteoporosis intervention guided by gene targets. While omics-based research into osteoporosis pathogenesis yields a plethora of potential gene targets for clinical transformation, identifying effective gene targets has posed challenges. Methods We first queried the omics data of osteoporosis clinical samples on PubMed, used International Mouse Phenotyping Consortium (IMPC) to screen differentially expressed genes, and conducted preliminary functional verification of candidate genes in human Saos2 cells through osteogenic differentiation and mineralization experiments. We then selected the candidate genes with the most significant effects on osteogenic differentiation and further verified the osteogenic differentiation and mineralization functions in mouse 3T3-E1 and bone marrow mesenchymal stem cells (BMSC). Finally, we used RNA-seq to explore the regulation of osteogenesis by the target gene. Results We identified PPP2R2A, RRBP1, HSPB6, SLC22A15, ADAMTS4, ATP8B1, CTNNB1, ROBO1, and EFR3B, which may contribute to osteoporosis. ROBO1 was the most significant regulator of osteogenesis in both human and mouse osteoblast. The inhibitory effect of Robo1 knockdown on osteogenic differentiation may be related to the activation of inflammatory signaling pathways. Conclusion Our study provides several novel molecular mechanisms involved in the pathogenesis of osteoporosis. ROBO1 is a potential target for osteoporosis intervention.
Collapse
Affiliation(s)
- Xiangzheng Zhang
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yike Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Zheng
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Wei
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ruizhi Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Keyu Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaocheng Zhai
- Division of Spine Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Youjia Xu
- The Osteoporosis Clinical Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
3
|
Bhadouria N, Holguin N. Osteoporosis treatments for intervertebral disc degeneration and back pain: a perspective. JBMR Plus 2024; 8:ziae048. [PMID: 38706880 PMCID: PMC11066806 DOI: 10.1093/jbmrpl/ziae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 05/07/2024] Open
Abstract
Low back pain derived from intervertebral disc (IVD) degeneration is a debilitating spinal condition that, despite its prevalence, does not have any intermediary guidelines for pharmacological treatment between palliative care and invasive surgery. The development of treatments for the IVD is complicated by the variety of resident cell types needed to maintain the regionally distinct structural properties of the IVD that permit the safe, complex motions of the spine. Osteoporosis of the spine increases the risk of vertebral bone fracture that can increase the incidence of back pain. Fortunately, there are a variety of pharmacological treatments for osteoporosis that target osteoblasts, osteoclasts and/or osteocytes to build bone and prevent vertebral fracture. Of particular note, clinical and preclinical studies suggest that commonly prescribed osteoporosis drugs like bisphosphonates, intermittent parathyroid hormone, anti-sclerostin antibody, selective estrogen receptor modulators and anti-receptor activator of nuclear factor-kappa B ligand inhibitor denosumab may also relieve back pain. Here, we cite clinical and preclinical studies and include unpublished data to support the argument that a subset of these therapeutics for osteoporosis may alleviate low back pain by also targeting the IVD.
Collapse
Affiliation(s)
- Neharika Bhadouria
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Nilsson Holguin
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
4
|
Tirandi A, Arboscello E, Ministrini S, Liberale L, Bonaventura A, Vecchié A, Bertolotto M, Giacobbe DR, Castellani L, Mirabella M, Minetti S, Bassetti M, Montecucco F, Carbone F. Early sclerostin assessment in frail elderly patients with sepsis: insights on short- and long-term mortality prediction. Intern Emerg Med 2023; 18:1509-1519. [PMID: 36943596 PMCID: PMC10412666 DOI: 10.1007/s11739-023-03223-w] [Citation(s) in RCA: 4] [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: 11/15/2022] [Accepted: 02/08/2023] [Indexed: 03/23/2023]
Abstract
Unmet needs challenge clinical management of sepsis especially concerning patient profiling, enhancing recovery, and long-term sequelae. Here, we preliminarily focused on sclerostin (SOST) as a candidate biomarker to encompass such a broad range of clinical needs related to sepsis. Seventy-three septic patients were enrolled at internal medicine wards between January 2017 and December 2019 in this pilot study. Clinical examination and blood sample analyses were collected at enrollment and after 7 and 14 days. SOST levels were assessed on serum by ELISA. Thirty-day mortality was set as primary outcome. In-hospital and long-term mortality (2.5 years of median follow-up) were assessed as secondary outcomes. Patients were frail, elderly, and heterogeneous in terms of comorbidity burden. SOST levels were associated with age, cardiovascular comorbidities, and time to early death (30 days). When regression models were built, SOST displayed a high predictive value toward 30-day mortality (OR 13.459 with 95% CI 1.226-148.017) with ever better performance than validated scoring scales for critical ill patients. Such a predictive value of SOST was further confirmed for in-hospital (HR 10.089 with 95% CI 1.375-74.013) and long-term mortality (HR 5.061 with 95% CI 1.379-18.570). SOST levels generally decreased over 7 to 14 days after enrollment (p for trend < 0.001). The degree of this variation further predicted long-term mortality (HR for Δ SOST T0-day 14: 1.006 with 95% CI 1.001-1.011). Our results suggest a role for SOST in both short- and long-time prediction of worse outcome in septic elderly admitted to internal medicine wards.
Collapse
Affiliation(s)
- Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Eleonora Arboscello
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, Università Degli Studi di Perugia, piazzale Gambuli 1, 06129, Perugia, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy
| | - Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | | | - Maria Bertolotto
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Daniele Roberto Giacobbe
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Luca Castellani
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy
| | - Michele Mirabella
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy
| | - Silvia Minetti
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
| | - Matteo Bassetti
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, 10 Largo Rosanna Benzi, 16132, Genoa, Italy.
| |
Collapse
|
5
|
Kim SP, Seward AH, Garcia-Diaz J, Alekos N, Gould NR, Aja S, Stains JP, Wolfgang MJ, Riddle RC. Peroxisome proliferator activated receptor-γ in osteoblasts controls bone formation and fat mass by regulating sclerostin expression. iScience 2023; 26:106999. [PMID: 37534168 PMCID: PMC10391670 DOI: 10.1016/j.isci.2023.106999] [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: 03/20/2023] [Revised: 05/01/2023] [Accepted: 05/25/2023] [Indexed: 08/04/2023] Open
Abstract
The nuclear receptor peroxisome proliferator activated receptor-γ (PPARγ) is a key contributor to metabolic function via its adipogenic and insulin-sensitizing functions, but it has negative effects on skeletal homeostasis. Here, we questioned whether the skeletal and metabolic actions of PPARγ are linked. Ablating Pparg expression in osteoblasts and osteocytes produced a high bone mass phenotype, secondary to increased osteoblast activity, and a reduction in subcutaneous fat mass because of reduced fatty acid synthesis and increased fat oxidation. The skeletal and metabolic phenotypes in Pparg mutants proceed from the regulation of sclerostin production by PPARγ. Mutants exhibited reductions in skeletal Sost expression and serum sclerostin levels while increasing production normalized both phenotypes. Importantly, disrupting the production of sclerostin synergized with the insulin-sensitizing actions of a PPARγ agonist while preventing bone loss. These data suggest that modulating sclerostin action may prevent bone loss associated with anti-diabetic therapies and augment their metabolic actions.
Collapse
Affiliation(s)
- Soohyun P. Kim
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Avery H. Seward
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jean Garcia-Diaz
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nathalie Alekos
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicole R. Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Michael J. Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ryan C. Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Research and Development Service, Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Movérare-Skrtic S, Voelkl J, Nilsson KH, Nethander M, Luong TTD, Alesutan I, Li L, Wu J, Horkeby K, Lagerquist MK, Koskela A, Tuukkanen J, Tobias JH, Lerner UH, Henning P, Ohlsson C. B4GALNT3 regulates glycosylation of sclerostin and bone mass. EBioMedicine 2023; 91:104546. [PMID: 37023531 PMCID: PMC10102813 DOI: 10.1016/j.ebiom.2023.104546] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Global sclerostin inhibition reduces fracture risk efficiently but has been associated with cardiovascular side effects. The strongest genetic signal for circulating sclerostin is in the B4GALNT3 gene region, but the causal gene is unknown. B4GALNT3 expresses the enzyme beta-1,4-N-acetylgalactosaminyltransferase 3 that transfers N-acetylgalactosamine onto N-acetylglucosaminebeta-benzyl on protein epitopes (LDN-glycosylation). METHODS To determine if B4GALNT3 is the causal gene, B4galnt3-/- mice were developed and serum levels of total sclerostin and LDN-glycosylated sclerostin were analysed and mechanistic studies were performed in osteoblast-like cells. Mendelian randomization was used to determine causal associations. FINDINGS B4galnt3-/- mice had higher circulating sclerostin levels, establishing B4GALNT3 as a causal gene for circulating sclerostin levels, and lower bone mass. However, serum levels of LDN-glycosylated sclerostin were lower in B4galnt3-/- mice. B4galnt3 and Sost were co-expressed in osteoblast-lineage cells. Overexpression of B4GALNT3 increased while silencing of B4GALNT3 decreased the levels of LDN-glycosylated sclerostin in osteoblast-like cells. Mendelian randomization demonstrated that higher circulating sclerostin levels, genetically predicted by variants in the B4GALNT3 gene, were causally associated with lower BMD and higher risk of fractures but not with higher risk of myocardial infarction or stroke. Glucocorticoid treatment reduced B4galnt3 expression in bone and increased circulating sclerostin levels and this may contribute to the observed glucocorticoid-induced bone loss. INTERPRETATION B4GALNT3 is a key factor for bone physiology via regulation of LDN-glycosylation of sclerostin. We propose that B4GALNT3-mediated LDN-glycosylation of sclerostin may be a bone-specific osteoporosis target, separating the anti-fracture effect of global sclerostin inhibition, from indicated cardiovascular side effects. FUNDING Found in acknowledgements.
Collapse
Affiliation(s)
- Sofia Movérare-Skrtic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria; Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Karin H Nilsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Maria Nethander
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Trang Thi Doan Luong
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Lei Li
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jianyao Wu
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin Horkeby
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marie K Lagerquist
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Jon H Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, and Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ulf H Lerner
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
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: 12] [Impact Index Per Article: 6.0] [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
|
Semenistaja S, Skuja S, Kadisa A, Groma V. Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk. Int J Mol Sci 2023; 24:4120. [PMID: 36835530 PMCID: PMC9964755 DOI: 10.3390/ijms24044120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive, severely debilitating, and multifactorial joint disease that is recognized as the most common type of arthritis. During the last decade, it shows an incremental global rise in prevalence and incidence. The interaction between etiologic factors that mediate joint degradation has been explored in numerous studies. However, the underlying processes that induce OA remain obscure, largely due to the variety and complexity of these mechanisms. During synovial joint dysfunction, the osteochondral unit undergoes cellular phenotypic and functional alterations. At the cellular level, the synovial membrane is influenced by cartilage and subchondral bone cleavage fragments and extracellular matrix (ECM) degradation products from apoptotic and necrotic cells. These "foreign bodies" serve as danger-associated molecular patterns (DAMPs) that trigger innate immunity, eliciting and sustaining low-grade inflammation in the synovium. In this review, we explore the cellular and molecular communication networks established between the major joint compartments-the synovial membrane, cartilage, and subchondral bone of normal and OA-affected joints.
Collapse
Affiliation(s)
- Sofija Semenistaja
- Department of Doctoral Studies, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Sandra Skuja
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Anda Kadisa
- Department of Internal Diseases, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Valerija Groma
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
| |
Collapse
|
9
|
O'Donohue AK, Xiao Y, Lee LR, Schofield T, Cheng TL, Munns CF, Baldock PA, Schindeler A. Targeted postnatal knockout of Sclerostin using a bone-targeted adeno-associated viral vector increases bone anabolism and decreases canalicular density. Bone 2023; 167:116636. [PMID: 36462771 DOI: 10.1016/j.bone.2022.116636] [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: 06/06/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
PURPOSE The creation of murine gene knockout models to study bone gene functions often requires the resource intensive crossbreeding of Cre transgenic and gene-floxed strains. The developmental versus postnatal roles of genes can be difficult to discern in such models. For example, embryonic deletion of the Sclerostin (Sost) gene establishes a high-bone mass phenotype in neonatal mice that may impact on future bone growth. To generate a postnatal skeletal knockout of Sost in adult mice, this study used a single injection of a bone-targeted recombinant adeno-associated virus (rAAV) vector. METHODS 8-week-old Sostflox/flox mice were injected with saline (control) or a single injection containing 5 × 1011 vg AAV8-Sp7-Cre vector. Ai9 fluorescent Cre reporter mice were dosed in parallel to confirm targeting efficiency. After 6 weeks, detailed bone analysis was performed via microCT, biomechanical testing, and bone histology on vertebral and long bone specimens. RESULTS The AAV8-Sp7-Cre vector induced widespread persistent recombination in the bone compartment. Regional microCT analyses revealed significant increases in bone with vector treatment. In the L3 vertebrae, Sostflox/flox:AAV-Cre showed a 22 % increase in bone volume and 21 % in trabecular bone fraction compared to controls; this translated to a 17 % increase in compressive strength. In the tibiae, Sostflox/flox:AAV-Cre led to small but statistically significant increases in cortical bone volume and thickness. These were consistent with a 25 % increase in mineral apposition rate, but this did not translate into increased four-point bending strength. Ploton silver nitrate stain on histological sections revealed an unexpected increase in canalicular density associated with Sost ablation. CONCLUSION This report demonstrates a proof-of-concept that the AAV8-Sp7-Cre vector can efficiently produce postnatal skeletal knockout mice using gene-floxed strains. This technology has the potential for broad utility in the bone field with existing conditional lines. These data also confirm an important postnatal role for Sost in regulating bone homeostasis, consistent with prior studies using neutralizing Sclerostin antibodies, and highlights a novel role of Sost in canalicular remodeling.
Collapse
Affiliation(s)
- Alexandra K O'Donohue
- Bioengineering & Molecular Medicine Laboratory, The Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, NSW, Australia; The Children's Hospital at Westmead Clinical School, The University of Sydney, Camperdown, NSW, Australia
| | - Ya Xiao
- Bone Division, Garvan Institute for Medical Research, Darlinghurst, NSW, Australia
| | - Lucinda R Lee
- Bioengineering & Molecular Medicine Laboratory, The Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, NSW, Australia; The Children's Hospital at Westmead Clinical School, The University of Sydney, Camperdown, NSW, Australia
| | - Timothy Schofield
- Bioengineering & Molecular Medicine Laboratory, The Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, NSW, Australia; The Children's Hospital at Westmead Clinical School, The University of Sydney, Camperdown, NSW, Australia
| | - Tegan L Cheng
- University of Sydney School of Health Sciences, University of Sydney, Camperdown, NSW, Australia; Engineering Prototypes & Implants for Children (EPIC) Lab, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Craig F Munns
- Department of Endocrinology and Diabetes, Queensland Children's Hospital, Brisbane, QLD, Australia; Child Health Research Centre and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Paul A Baldock
- Bone Division, Garvan Institute for Medical Research, Darlinghurst, NSW, Australia
| | - Aaron Schindeler
- Bioengineering & Molecular Medicine Laboratory, The Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, NSW, Australia; The Children's Hospital at Westmead Clinical School, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
10
|
Riddle RC. Endocrine Functions of Sclerostin. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2023; 28:10.1016/j.coemr.2022.100433. [PMID: 36713826 PMCID: PMC9881182 DOI: 10.1016/j.coemr.2022.100433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Sclerostin, the product of the SOST gene has primarily been studied for its profound impact on bone mass. By interacting with LRP5 and LRP6, the glycoprotein suppresses the propagation of Wnt signals to β-catenin and thereby suppresses new bone formation. In this review, we discuss emerging data which suggest that sclerostin also acts outside the skeleton to influence metabolism. In humans, serum sclerostin levels are associated with body mass index and indices of metabolic function. Likewise, genetic mouse models of Sost gene deficiency indicate sclerostin influences adipocyte development and insulin signaling. These data raise the possibility that sclerostin neutralization may be effective at treating two epidemic conditions: osteoporosis and obesity.
Collapse
Affiliation(s)
- Ryan C. Riddle
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Research and Development Service, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA.,Address Correspondence to: Ryan C. Riddle, Ph.D., Department of Orthopaedics, University of Maryland School of Medicine, 660 W. Redwood Street, Room 592, Baltimore, MD 21201, USA, , Ph: 410-706-0422
| |
Collapse
|
11
|
Ren Y, Bäcker H, Müller M, Kienzle A. The role of myeloid derived suppressor cells in musculoskeletal disorders. Front Immunol 2023; 14:1139683. [PMID: 36936946 PMCID: PMC10020351 DOI: 10.3389/fimmu.2023.1139683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The immune system is closely linked to bone homeostasis and plays a pivotal role in several pathological and inflammatory conditions. Through various pathways it modulates various bone cells and subsequently sustains the physiological bone metabolism. Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous immature myeloid-derived cells that can exert an immunosuppressive function through a direct cell-to-cell contact, secretion of anti-inflammatory cytokines or specific exosomes. These cells mediate the innate immune response to chronic stress on the skeletal system. In chronic inflammation, MDSCs act as an inner offset to rebalance overactivation of the immune system. Moreover, they have been found to be involved in processes responsible for bone remodeling in different musculoskeletal disorders, autoimmune diseases, infection, and cancer. These cells can not only cause bone erosion by differentiating into osteoclasts, but also alleviate the immune reaction, subsequently leading to long-lastingly impacted bone remodeling. In this review, we discuss the impact of MDSCs on the bone metabolism under several pathological conditions, the involved modulatory pathways as well as potential therapeutic targets in MDSCs to improve bone health.
Collapse
Affiliation(s)
- Yi Ren
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Henrik Bäcker
- Department of Orthopedics, Auckland City Hospital, Auckland, New Zealand
| | - Michael Müller
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Arne Kienzle
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health, Charité — Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Arne Kienzle,
| |
Collapse
|
12
|
Zhang Y, Zhao Y, Xie Z, Li M, Liu Y, Tu X. Activating Wnt/β-Catenin Signaling in Osteocytes Promotes Osteogenic Differentiation of BMSCs through BMP-7. Int J Mol Sci 2022; 23:ijms232416045. [PMID: 36555684 PMCID: PMC9785209 DOI: 10.3390/ijms232416045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Bone formation is critically needed in orthopedic clinical practice. We found that, bone morphogenetic protein-7 (BMP-7) gene expression was significantly increased in fractured mice, which activates canonical Wnt signaling exclusively in osteocytes. Wnt and BMP signaling appear to exhibit synergistic or antagonistic effects in different kinds of cells. However, the communication between Wnt/β-catenin signaling and BMP signaling in osteocytes is almost unknown. Our study verified in vitro that BMP-7 expression was significantly increased when Wnt signaling was activated in osteocytes. Next, BMP-7 in osteocytes was overexpressed using an adenovirus, the osteogenesis of bone marrow stem cells (BMSCs) was enhanced, when cocultured with osteocytes. On the contrary, BMP-7 in osteocytes was silenced using an adenovirus, the osteogenesis of bone marrow stem cells (BMSCs) was weakened. In addition, the osteogenesis of BMSCs was no longer promoted by Wnt-activated osteocytes when BMP-7 was silenced. Therefore, the results showed that BMP-7 mediated the anabolic actions of Wnt/β-catenin signaling in osteocytes. Our study provides new evidence for the clinical application of BMP-7-overexpressed osteocytes.
Collapse
Affiliation(s)
- Yining Zhang
- Correspondence: (Y.Z.); (X.T.); Tel.: +86-23-6365-1934 (X.T.)
| | | | | | | | | | - Xiaolin Tu
- Correspondence: (Y.Z.); (X.T.); Tel.: +86-23-6365-1934 (X.T.)
| |
Collapse
|
13
|
Kim MJ, Valderrábano RJ, Wu JY. Osteoblast Lineage Support of Hematopoiesis in Health and Disease. J Bone Miner Res 2022; 37:1823-1842. [PMID: 35983701 PMCID: PMC11346465 DOI: 10.1002/jbmr.4678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/21/2022] [Accepted: 08/13/2022] [Indexed: 11/06/2022]
Abstract
In mammals, hematopoiesis migrates to the bone marrow during embryogenesis coincident with the appearance of mineralized bone, where hematopoietic stem cells (HSCs) and their progeny are maintained by the surrounding microenvironment or niche, and sustain the entirety of the hematopoietic system. Genetic manipulation of niche factors and advances in cell lineage tracing techniques have implicated cells of both hematopoietic and nonhematopoietic origin as important regulators of hematopoiesis in health and disease. Among them, cells of the osteoblast lineage, from stromal skeletal stem cells to matrix-embedded osteocytes, are vital niche residents with varying capacities for hematopoietic support depending on stage of differentiation. Here, we review populations of osteoblasts at differing stages of differentiation and summarize the current understanding of the role of the osteoblast lineage in supporting hematopoiesis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Matthew J Kim
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rodrigo J Valderrábano
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joy Y Wu
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
14
|
Kroon T, Bhadouria N, Niziolek P, Edwards D, Choi R, Clinkenbeard EL, Robling A, Holguin N. Suppression of Sost/Sclerostin and Dickkopf-1 Augment Intervertebral Disc Structure in Mice. J Bone Miner Res 2022; 37:1156-1169. [PMID: 35278242 PMCID: PMC9320845 DOI: 10.1002/jbmr.4546] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 11/06/2022]
Abstract
Intervertebral disc (IVD) degeneration is a leading cause of low back pain, characterized by accelerated extracellular matrix breakdown and IVD height loss, but there is no approved pharmacological therapeutic. Deletion of Wnt ligand competitor Lrp5 induces IVD degeneration, suggesting that Wnt signaling is essential for IVD homeostasis. Therefore, the IVD may respond to neutralization of Wnt ligand competitors sost(gene)/sclerostin(protein) and/or dickkopf-1 (dkk1). Anti-sclerostin antibody (scl-Ab) is an FDA-approved bone therapeutic that activates Wnt signaling. We aimed to (i) determine if pharmacological neutralization of sclerostin, dkk1, or their combination would stimulate Wnt signaling and augment IVD structure and (ii) determine the prolonged adaptation of the IVD to global, persistent deletion of sost. Nine-week-old C57Bl/6J female mice (n = 6-7/group) were subcutaneously injected 2×/week for 5.5 weeks with scl-Ab (25 mg/kg), dkk1-Ab (25 mg/kg), 3:1 scl-Ab/dkk1-Ab (18.75:6.25 mg/kg), or vehicle (veh). Separately, IVD of sost KO and wild-type (WT) mice (n = 8/group) were harvested at 16 weeks of age. First, compared with vehicle, injection of scl-Ab, dkk1-Ab, and 3:1 scl-Ab/dkk1-Ab similarly increased lumbar IVD height and β-catenin gene expression. Despite these similarities, only injection of scl-Ab alone strengthened IVD mechanical properties and decreased heat shock protein gene expressions. Genetically and compared with WT, sost KO enlarged IVD height, increased proteoglycan staining, and imbibed IVD hydration. Notably, persistent deletion of sost was compensated by upregulation of dkk1, which consequently reduced the cell nuclear fraction for Wnt signaling co-transcription factor β-catenin in the IVD. Lastly, RNA-sequencing pathway analysis confirmed the compensatory suppression of Wnt signaling and revealed a reduction of cellular stress-related pathways. Together, suppression of sost/sclerostin or dkk1 each augmented IVD structure by stimulating Wnt signaling, but scl-Ab outperformed dkk1-Ab in strengthening the IVD. Ultimately, postmenopausal women prescribed scl-Ab injections to prevent vertebral fracture may also benefit from a restoration of IVD height and health. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tori Kroon
- Department of Biomedical EngineeringIUPUIIndianapolisINUSA
| | - Neharika Bhadouria
- Department of Mechanical EngineeringPurdue UniversityWest LafayetteINUSA
| | | | - Daniel Edwards
- Indiana Center of Musculoskeletal HealthIndianapolisINUSA
| | - Roy Choi
- Department for Anatomy and Cell BiologyIUPUIIndianapolisINUSA
| | | | - Alexander Robling
- Indiana Center of Musculoskeletal HealthIndianapolisINUSA
- Department for Anatomy and Cell BiologyIUPUIIndianapolisINUSA
| | - Nilsson Holguin
- Indiana Center of Musculoskeletal HealthIndianapolisINUSA
- Department for Anatomy and Cell BiologyIUPUIIndianapolisINUSA
- Department of Mechanical and Energy EngineeringIUPUIIndianapolisINUSA
| |
Collapse
|
15
|
Chicana B, Abbasizadeh N, Burns C, Taglinao H, Spencer JA, Manilay JO. Deletion of Vhl in Dmp1-Expressing Cells Causes Microenvironmental Impairment of B Cell Lymphopoiesis. Front Immunol 2022; 13:780945. [PMID: 35250971 PMCID: PMC8889104 DOI: 10.3389/fimmu.2022.780945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/11/2022] [Indexed: 12/25/2022] Open
Abstract
The contributions of skeletal cells to the processes of B cell development in the bone marrow (BM) have not been completely described. The von-Hippel Lindau protein (VHL) plays a key role in cellular responses to hypoxia. Previous work showed that Dmp1-Cre;Vhl conditional knockout mice (VhlcKO), which deletes Vhl in subsets of mesenchymal stem cells, late osteoblasts and osteocytes, display dysregulated bone growth and reduction in B cells. Here, we investigated the mechanisms underlying the B cell defects using flow cytometry and high-resolution imaging. In the VhlcKO BM, B cell progenitors were increased in frequency and number, whereas Hardy Fractions B-F were decreased. VhlcKO Fractions B-C cells showed increased apoptosis and quiescence. Reciprocal BM chimeras confirmed a B cell-extrinsic source of the VhlcKO B cell defects. In support of this, VhlcKO BM supernatant contained reduced CXCL12 and elevated EPO levels. Intravital and ex vivo imaging revealed VhlcKO BM blood vessels with increased diameter, volume, and a diminished blood-BM barrier. Staining of VhlcKO B cells with an intracellular hypoxic marker indicated the natural existence of distinct B cell microenvironments that differ in local oxygen tensions and that the B cell developmental defects in VhlcKO BM are not initiated by hypoxia. Our studies identify novel mechanisms linking altered bone homeostasis with drastic BM microenvironmental changes that dysregulate B cell development.
Collapse
Affiliation(s)
- Betsabel Chicana
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Christian Burns
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Hanna Taglinao
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Joel A Spencer
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States.,Bioengineering Graduate Program, University of California, Merced, Merced, CA, United States
| | - Jennifer O Manilay
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| |
Collapse
|
16
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
17
|
Kim SP, Da H, Wang L, Taketo MM, Wan M, Riddle RC. Bone-derived sclerostin and Wnt/β-catenin signaling regulate PDGFRα + adipoprogenitor cell differentiation. FASEB J 2021; 35:e21957. [PMID: 34606641 PMCID: PMC8496915 DOI: 10.1096/fj.202100691r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022]
Abstract
The Wnt signaling antagonist, sclerostin, is a potent suppressor of bone acquisition that also mediates endocrine communication between bone and adipose. As a result, Sost-/- mice exhibit dramatic increases in bone formation but marked decreases in visceral and subcutaneous adipose that are secondary to alterations in lipid synthesis and utilization. While interrogating the mechanism by which sclerostin influences adipocyte metabolism, we observed paradoxical increases in the adipogenic potential and numbers of CD45- :Sca1+ :PDGFRα+ adipoprogenitors in the stromal vascular compartment of fat pads isolated from male Sost-/- mice. Lineage tracing studies indicated that sclerostin deficiency blocks the differentiation of PDGFRα+ adipoprogenitors to mature adipocytes in association with increased Wnt/β-catenin signaling. Importantly, osteoblast/osteocyte-specific Sost gene deletion mirrors the accumulation of PDGFRα+ adipoprogenitors, reduction in fat mass, and improved glucose metabolism evident in Sost-/- mice. These data indicate that bone-derived sclerostin regulates multiple facets of adipocyte physiology ranging from progenitor cell commitment to anabolic metabolism.
Collapse
Affiliation(s)
- Soohyun P Kim
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Da
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Makoto M Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Research and Development Service, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Sclerostin Depletion Induces Inflammation in the Bone Marrow of Mice. Int J Mol Sci 2021; 22:ijms22179111. [PMID: 34502021 PMCID: PMC8431516 DOI: 10.3390/ijms22179111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 01/25/2023] Open
Abstract
Romosozumab, a humanized monoclonal antibody specific for sclerostin (SOST), has been approved for treatment of postmenopausal women with osteoporosis at a high risk for fracture. Previous work in sclerostin global knockout (Sost-/-) mice indicated alterations in immune cell development in the bone marrow (BM), which could be a possible side effect in romosozumab-treated patients. Here, we examined the effects of short-term sclerostin depletion in the BM on hematopoiesis in young mice receiving sclerostin antibody (Scl-Ab) treatment for 6 weeks, and the effects of long-term Sost deficiency on wild-type (WT) long-term hematopoietic stem cells transplanted into older cohorts of Sost-/- mice. Our analyses revealed an increased frequency of granulocytes in the BM of Scl-Ab-treated mice and WT→Sost-/- chimeras, indicating myeloid-biased differentiation in Sost-deficient BM microenvironments. This myeloid bias extended to extramedullary hematopoiesis in the spleen and was correlated with an increase in inflammatory cytokines TNFα, IL-1α, and MCP-1 in Sost-/- BM serum. Additionally, we observed alterations in erythrocyte differentiation in the BM and spleen of Sost-/- mice. Taken together, our current study indicates novel roles for Sost in the regulation of myelopoiesis and control of inflammation in the BM.
Collapse
|
19
|
Wang X, Li Z, Wang C, Bai H, Wang Z, Liu Y, Bao Y, Ren M, Liu H, Wang J. Enlightenment of Growth Plate Regeneration Based on Cartilage Repair Theory: A Review. Front Bioeng Biotechnol 2021; 9:654087. [PMID: 34150725 PMCID: PMC8209549 DOI: 10.3389/fbioe.2021.654087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/10/2021] [Indexed: 01/21/2023] Open
Abstract
The growth plate (GP) is a cartilaginous region situated between the epiphysis and metaphysis at the end of the immature long bone, which is susceptible to mechanical damage because of its vulnerable structure. Due to the limited regeneration ability of the GP, current clinical treatment strategies (e.g., bone bridge resection and fat engraftment) always result in bone bridge formation, which will cause length discrepancy and angular deformity, thus making satisfactory outcomes difficult to achieve. The introduction of cartilage repair theory and cartilage tissue engineering technology may encourage novel therapeutic approaches for GP repair using tissue engineered GPs, including biocompatible scaffolds incorporated with appropriate seed cells and growth factors. In this review, we summarize the physiological structure of GPs, the pathological process, and repair phases of GP injuries, placing greater emphasis on advanced tissue engineering strategies for GP repair. Furthermore, we also propose that three-dimensional printing technology will play a significant role in this field in the future given its advantage of bionic replication of complex structures. We predict that tissue engineering strategies will offer a significant alternative to the management of GP injuries.
Collapse
Affiliation(s)
- Xianggang Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Chenyu Wang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, China
| | - Haotian Bai
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Zhonghan Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yuzhe Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yirui Bao
- Department of Orthopedics, Chinese PLA 965 Hospital, Jilin, China
| | - Ming Ren
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| |
Collapse
|
20
|
Baroi S, Czernik PJ, Chougule A, Griffin PR, Lecka-Czernik B. PPARG in osteocytes controls sclerostin expression, bone mass, marrow adiposity and mediates TZD-induced bone loss. Bone 2021; 147:115913. [PMID: 33722775 PMCID: PMC8076091 DOI: 10.1016/j.bone.2021.115913] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 01/07/2023]
Abstract
The peroxisome proliferator activated receptor gamma (PPARG) nuclear receptor regulates energy metabolism and insulin sensitivity. In this study, we present novel evidence for an essential role of PPARG in the regulation of osteocyte function, and support for the emerging concept of the conjunction between regulation of energy metabolism and bone mass. We report that PPARG is essential for sclerostin production, a recently approved target to treat osteoporosis. Our mouse model of osteocyte-specific PPARG deletion (Dmp1CrePparγflfl or γOTKO) is characterized with increased bone mass and reduced bone marrow adiposity, which is consistent with upregulation of WNT signaling and increased bone forming activity of endosteal osteoblasts. An analysis of osteocytes derived from γOTKO and control mice showed an excellent correlation between PPARG and SOST/sclerostin at the transcript and protein levels. The 8 kb sequence upstream of Sost gene transcription start site possesses multiple PPARG binding elements (PPREs) with at least two of them binding PPARG with dynamics reflecting its activation with full agonist rosiglitazone and correlating with increased levels of Sost transcript and sclerostin protein expression (Pearson's r = 0.991, p = 0.001). Older γOTKO female mice are largely protected from TZD-induced bone loss providing proof of concept that PPARG in osteocytes can be pharmacologically targeted. These findings demonstrate that transcriptional activities of PPARG are essential for sclerostin expression in osteocytes and support consideration of targeting PPARG activities with selective modulators to treat osteoporosis.
Collapse
Affiliation(s)
- Sudipta Baroi
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Piotr J Czernik
- Department of Physiology and Pharmacology, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Amit Chougule
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Patrick R Griffin
- The Scripps Research Institute, Jupiter, FL, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America; Department of Physiology and Pharmacology, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America; Center for Diabetes and Endocrine Research, University of Toledo, College of Medicine and Life Sciences, Toledo, OH, United States of America.
| |
Collapse
|
21
|
Tsukasaki M, Asano T, Muro R, Huynh NCN, Komatsu N, Okamoto K, Nakano K, Okamura T, Nitta T, Takayanagi H. OPG Production Matters Where It Happened. Cell Rep 2021; 32:108124. [PMID: 32905763 DOI: 10.1016/j.celrep.2020.108124] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/05/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
Osteoprotegerin (OPG) is a circulating decoy receptor for RANKL, a multifunctional cytokine essential for the differentiation of tissue-specific cells in bone and immune systems such as osteoclasts, medullary thymic epithelial cells (mTECs), and intestinal microfold cells (M cells). However, it is unknown whether OPG functions only at the production site or circulates to other tissues acting in an endocrine fashion. Here we explore the cellular source of OPG by generating OPG-floxed mice and show that locally produced OPG, rather than circulating OPG, is crucial for bone and immune homeostasis. Deletion of OPG in osteoblastic cells leads to severe osteopenia without affecting serum OPG. Deletion of locally produced OPG increases mTEC and M cell numbers while retaining the normal serum OPG level. This study shows that OPG limits its functions within the tissue where it was produced, illuminating the importance of local regulation of the RANKL system.
Collapse
Affiliation(s)
- Masayuki Tsukasaki
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Tatsuo Asano
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Nam Cong-Nhat Huynh
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, 162-8655 Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, 162-8655 Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan.
| |
Collapse
|
22
|
Endocrine role of bone in the regulation of energy metabolism. Bone Res 2021; 9:25. [PMID: 34016950 PMCID: PMC8137703 DOI: 10.1038/s41413-021-00142-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Bone mainly functions as a supportive framework for the whole body and is the major regulator of calcium homeostasis and hematopoietic function. Recently, an increasing number of studies have characterized the significance of bone as an endocrine organ, suggesting that bone-derived factors regulate local bone metabolism and metabolic functions. In addition, these factors can regulate global energy homeostasis by altering insulin sensitivity, feeding behavior, and adipocyte commitment. These findings may provide a new pathological mechanism for related metabolic diseases or be used in the diagnosis, treatment, and prevention of metabolic diseases such as osteoporosis, obesity, and diabetes mellitus. In this review, we summarize the regulatory effect of bone and bone-derived factors on energy metabolism and discuss directions for future research.
Collapse
|
23
|
Abstract
Skeletal integrity is maintained by a meticulous balance between bone resorption and bone formation, and recent studies have revealed the essential role of canonical Wnt signaling pathways in maintaining skeletal homeostasis. The SOST gene, which encodes sclerostin, a member of Dan family glycoproteins, was originally identified as the gene responsible for two sclerosing bone dysplasias, sclerosteosis and van Buchem disease. Sclerostin is highly expressed by osteocytes, negatively regulates canonical Wnt signaling pathways by binding to low-density lipoprotein receptor-related protein (LRP) 5/6, and suppresses osteoblast differentiation and/or function. Romosozumab, a specific anti-sclerostin antibody, inhibits sclerostin-LRP5/6 interactions and indirectly activates canonical Wnt signaling pathways and bone formation. This review focuses on the mechanism of action of sclerostin and summarizes clinical studies that demonstrated the efficacy of romosozumab to increase bone mineral density and reduce osteoporotic fractures, as well as its cardiovascular safety.
Collapse
Affiliation(s)
- Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
24
|
Activation of creER recombinase in the mouse calvaria induces local recombination without effects on distant skeletal segments. Sci Rep 2021; 11:8214. [PMID: 33859263 PMCID: PMC8050205 DOI: 10.1038/s41598-021-87611-2] [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: 11/18/2020] [Accepted: 03/31/2021] [Indexed: 02/02/2023] Open
Abstract
Conditional creER-mediated gene inactivation or gene induction has emerged as a robust tool for studying gene functions in mouse models of tissue development, homeostasis, and regeneration. Here, we present a method to conditionally induce cre recombination in the mouse calvarial bone while avoiding systemic recombination in distal bones. To test our method, we utilized Prx1creER-egfp;td-Tomato mice and delivered 4-hydroxytamoxifen (4-OHT) to the mouse calvaria, subperiosteally. First, we showed that two calvaria subperiosteal injections of 10 µg of 4-OHT (3.3 mg of 4-OHT/kg of body weight) can induce local recombination as efficiently as two intraperitoneal systemic injections of 200 μg of tamoxifen (70 mg of tamoxifen/kg of body weight). Then, we studied the recombination efficiency of various subperiosteal calvaria dosages and found that two subperiosteal injections of 5 µg 4-OHT (1.65 mg of 4-OHT/kg of body weight) uphold the same recombination efficiency observed with higher dosages. Importantly, the result indicated that the low dosage does not induce significant systemic recombination in remote skeletal tissues. With the proposed local low dosage protocol, the recombination efficiency at the injection site (calvarial bone) reached 94%, while the recombination efficiency at the mandible and the digits was as low as the efficiency measured in control animals.
Collapse
|
25
|
Biosse Duplan M, Dambroise E, Estibals V, Veziers J, Guicheux J, Legeai-Mallet L. An Fgfr3-activating mutation in immature murine osteoblasts affects the appendicular and craniofacial skeleton. Dis Model Mech 2021; 14:dmm048272. [PMID: 33737326 PMCID: PMC8084574 DOI: 10.1242/dmm.048272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Achondroplasia (ACH), the most common form of dwarfism, is caused by a missense mutation in the gene coding for fibroblast growth factor receptor 3 (FGFR3). The resulting increase in FGFR3 signaling perturbs the proliferation and differentiation of chondrocytes (CCs), alters the process of endochondral ossification and thus reduces bone elongation. Increased FGFR3 signaling in osteoblasts (OBs) might also contribute to bone anomalies in ACH. In the present study of a mouse model of ACH, we sought to determine whether FGFR3 overactivation in OBs leads to bone modifications. The model carries an Fgfr3-activating mutation (Fgfr3Y367C/+) that accurately mimics ACH; we targeted the mutation to either immature OBs and hypertrophic CCs or to mature OBs by using the Osx-cre and collagen 1α1 (2.3 kb Col1a1)-cre mouse strains, respectively. We observed that Fgfr3 activation in immature OBs and hypertrophic CCs (Osx-Fgfr3) not only perturbed the hypertrophic cells of the growth plate (thus affecting long bone growth) but also led to osteopenia and low cortical thickness in long bones in adult (3-month-old) mice but not growing (3-week-old) mice. Importantly, craniofacial membranous bone defects were present in the adult mice. In contrast, activation of Fgfr3 in mature OBs (Col1-Fgfr3) had very limited effects on skeletal shape, size and micro-architecture. In vitro, we observed that Fgfr3 activation in immature OBs was associated with low mineralization activity. In conclusion, immature OBs appear to be affected by Fgfr3 overactivation, which might contribute to the bone modifications observed in ACH independently of CCs.
Collapse
Affiliation(s)
- Martin Biosse Duplan
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
- Service de Médecine Bucco-Dentaire, Hôpital Bretonneau, AP-HP, Paris 75018, France
| | - Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| | - Valentin Estibals
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| | - Joelle Veziers
- Inserm, UMR 1229, RMeS – Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, F-44042, France
- SC3M, SFR Santé F. Bonamy, FED 4203, UMS Inserm 016, CNRS 3556, Nantes F-44042, France
- CHU Nantes, PHU4 OTONN, Nantes, F-44093, France
| | - Jérome Guicheux
- Inserm, UMR 1229, RMeS – Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, F-44042, France
- SC3M, SFR Santé F. Bonamy, FED 4203, UMS Inserm 016, CNRS 3556, Nantes F-44042, France
- CHU Nantes, PHU4 OTONN, Nantes, F-44093, France
| | - Laurence Legeai-Mallet
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| |
Collapse
|
26
|
Sun M, Chen Z, Wu X, Yu Y, Wang L, Lu A, Zhang G, Li F. The Roles of Sclerostin in Immune System and the Applications of Aptamers in Immune-Related Research. Front Immunol 2021; 12:602330. [PMID: 33717084 PMCID: PMC7946814 DOI: 10.3389/fimmu.2021.602330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/14/2021] [Indexed: 12/19/2022] Open
Abstract
Wnt signaling is one of the fundamental pathways that play a major role in almost every aspect of biological systems. In addition to the well-known influence of Wnt signaling on bone formation, its essential role in the immune system also attracted increasing attention. Sclerostin, a confirmed Wnt antagonist, is also proven to modulate the development and differentiation of normal immune cells, particularly B cells. Aptamers, single-stranded (ss) oligonucleotides, are capable of specifically binding to a variety of target molecules by virtue of their unique three-dimensional structures. With in-depth study of those functional nucleic acids, they have been gradually applied to diagnostic and therapeutic area in immune diseases due to their various advantages over antibodies. In this review, we focus on several issues including the roles of Wnt signaling and Wnt antagonist sclerostin in the immune system. For the sake of understanding, current examples of aptamers applications for the immune diseases are also discussed. At the end of this review, we propose our ideas for the future research directions.
Collapse
Affiliation(s)
- Meiheng Sun
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoqiu Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| |
Collapse
|
27
|
Liu L, Yuan Y, Zhang S, Xu J, Zou J. Osteoimmunological insights into the pathogenesis of ankylosing spondylitis. J Cell Physiol 2021; 236:6090-6100. [PMID: 33559242 DOI: 10.1002/jcp.30313] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 01/10/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Ankylosing spondylitis (AS) is inflammatory arthritis predominantly affecting the spine, which is involved in the disorders of both immune and skeletal systems. The exact pathogenesis of AS is not fully understood. Osteoimmunology is a new subject of study in inflammatory arthritis, in particular the pathogenic events involved in the cross-regulation of both skeletal and immune systems. In this review, we discuss osteoimmunological and pathological changes of AS in the spine that are characterized by altered osteogenesis and osteolytic bone destruction, accompanied by the changes of the immune system. It was revealed that bone cells like mesenchymal stem cells, osteoblast, and osteoclast in crossing talking with immune cells such as T cells, B cells coregulate to the pathogenesis of AS. Further, an array of cytokines and molecules expressed by both skeletal and immune systems contribute to these complex interplays. Understanding the cellular and molecular mechanisms underlying the pathogenesis of AS will lay a foundation for the exploration of the potential new treatment to AS.
Collapse
Affiliation(s)
- Lifei Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation, The People's Hospital of Liaoning Province, Shenyang, China
| | - Yu Yuan
- School of Sport and Health, Guangzhou Sport University, Guangzhou, China
| | - Shihua Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
28
|
Li H, Xiao Z, Quarles LD, Li W. Osteoporosis: Mechanism, Molecular Target and Current Status on Drug Development. Curr Med Chem 2021; 28:1489-1507. [PMID: 32223730 PMCID: PMC7665836 DOI: 10.2174/0929867327666200330142432] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/22/2022]
Abstract
CDATA[Osteoporosis is a pathological loss of bone mass due to an imbalance in bone remodeling where osteoclast-mediated bone resorption exceeds osteoblast-mediated bone formation resulting in skeletal fragility and fractures. Anti-resorptive agents, such as bisphosphonates and SERMs, and anabolic drugs that stimulate bone formation, including PTH analogues and sclerostin inhibitors, are current treatments for osteoporosis. Despite their efficacy, severe side effects and loss of potency may limit the long term usage of a single drug. Sequential and combinational use of current drugs, such as switching from an anabolic to an anti-resorptive agent, may provide an alternative approach. Moreover, there are novel drugs being developed against emerging new targets such as Cathepsin K and 17β-HSD2 that may have less side effects. This review will summarize the molecular mechanisms of osteoporosis, current drugs for osteoporosis treatment, and new drug development strategies.
Collapse
Affiliation(s)
- Hanxuan Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38165, USA
| | - L. Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38165, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| |
Collapse
|
29
|
Wang JS, Mazur CM, Wein MN. Sclerostin and Osteocalcin: Candidate Bone-Produced Hormones. Front Endocrinol (Lausanne) 2021; 12:584147. [PMID: 33776907 PMCID: PMC7988212 DOI: 10.3389/fendo.2021.584147] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/13/2021] [Indexed: 12/22/2022] Open
Abstract
In addition to its structural role, the skeleton serves as an endocrine organ that controls mineral metabolism and energy homeostasis. Three major cell types in bone - osteoblasts, osteoclasts, and osteocytes - dynamically form and maintain bone and secrete factors with systemic activity. Osteocalcin, an osteoblast-derived factor initially described as a matrix protein that regulates bone mineralization, has been suggested to be an osteoblast-derived endocrine hormone that regulates multiple target organs including pancreas, liver, muscle, adipose, testes, and the central and peripheral nervous system. Sclerostin is predominantly produced by osteocytes, and is best known as a paracrine-acting regulator of WNT signaling and activity of osteoblasts and osteoclasts on bone surfaces. In addition to this important paracrine role for sclerostin within bone, sclerostin protein has been noted to act at a distance to regulate adipocytes, energy homeostasis, and mineral metabolism in the kidney. In this article, we aim to bring together evidence supporting an endocrine function for sclerostin and osteocalcin, and discuss recent controversies regarding the proposed role of osteocalcin outside of bone. We summarize the current state of knowledge on animal models and human physiology related to the multiple functions of these bone-derived factors. Finally, we highlight areas in which future research is expected to yield additional insights into the biology of osteocalcin and sclerostin.
Collapse
Affiliation(s)
- Jialiang S. Wang
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Courtney M. Mazur
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marc N. Wein
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
- *Correspondence: Marc N. Wein,
| |
Collapse
|
30
|
Lee JY, Yang JY, Kim SW. Bone Lining Cells Could Be Sources of Bone Marrow Adipocytes. Front Endocrinol (Lausanne) 2021; 12:766254. [PMID: 34925236 PMCID: PMC8678613 DOI: 10.3389/fendo.2021.766254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recently, lineage-tracing studies demonstrated that parathyroid hormone and anti-sclerostin antibody (Scl-Ab) can convert bone lining cells (BLCs) into active osteoblasts. However, BLCs might also be differentiated into other lineages. Here we investigated whether BLCs could differentiate into bone marrow adipocytes (BMAds) and whether Scl-Ab could suppress this process. METHODS Dmp1-CreERt2:mTmG mice were injected with 0.5 mg of 4-hydroxytamoxifen once weekly from postnatal week 4 to week 8. The mice were treated with either vehicle or rosiglitazone for 8 weeks (weeks 12-20). Moreover, they were administered either vehicle or Scl-Ab (50 mg/kg) twice weekly for 4 weeks (weeks 16-20, N = 4-6/group). We chased the GFP+ cells from the endosteal surface to the bone marrow (BM) of the femur. Using immunohistochemical staining, the numbers of perilipin+ or GFP+/perilipin double+ cells in the BM were quantified. In addition, serum N-terminal propeptide of type I procollagen (P1NP) levels were measured at each time point, and bone mass was analyzed at 20 weeks using micro-computed tomography. RESULTS Scl-Ab administration significantly reversed the decreases in bone parameters induced by rosiglitazone. Plump GFP+ cells, presumably active osteoblasts, and extremely flat GFP+ cells, presumably BLCs, were present on the endosteal surface of the femur at 8 and 12 weeks, respectively, in line with prior findings. When we chased the GFP+ cells, rosiglitazone significantly increased the number of GFP/perilipin double+ BMAds compared to the effects of the vehicle (P < 0.001), and overlapping Scl-Ab administration decreased the number of GFP/perilipin double + BMAd compared to rosiglitazone alone (P < 0.001). In addition, we found that osteoblast lineage cells such as BLCs might express PPARγ on immunohistochemical staining. When rosiglitazone was administered to Rip-Cre:mTmG mice, GFP+ cells were not present on the endosteal surface or in the BM of the femur; however, they were present in the pancreas. CONCLUSION BLCs could be sources of BMAds, and rosiglitazone could stimulate the differentiation of osteoblast lineage cells into BMAds. Suppression of the differentiation of osteoblast lineage cells into BMAds might contribute to anabolic effects resulting from the pharmacologic inhibition of sclerostin.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Research and Experiment, Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| | - Jae-Yeon Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Research and Experiment, Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| | - Sang Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Division of Endocrinology and Metabolism, Seoul Metropolitan Government Boramae Medical Center, Seoul, South Korea
- *Correspondence: Sang Wan Kim,
| |
Collapse
|
31
|
Mendez ME, Murugesh DK, Sebastian A, Hum NR, McCloy SA, Kuhn EA, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Improves Post-Traumatic Osteoarthritis Outcomes in Mice. Int J Mol Sci 2020; 21:E6424. [PMID: 32899361 PMCID: PMC7503363 DOI: 10.3390/ijms21176424] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease characterized by the chronic and progressive degradation of articular cartilage. Post-traumatic OA (PTOA) is a secondary form of OA that develops in ~50% of cases of severe articular injury. Inflammation and re-occurring injury have been implicated as contributing to the progression of PTOA after the initial injury. However, there is very little known about external factors prior to injury that could affect the risk of PTOA development. To examine how the gut microbiome affects PTOA development we used a chronic antibiotic treatment regimen starting at weaning for six weeks prior to ACL rupture, in mice. A six-weeks post-injury histological examination showed more robust cartilage staining on the antibiotic (AB)-treated mice than the untreated controls (VEH), suggesting slower disease progression in AB cohorts. Injured joints also showed an increase in the presence of anti-inflammatory M2 macrophages in the AB group. Molecularly, the phenotype correlated with a significantly lower expression of inflammatory genes Tlr5, Ccl8, Cxcl13, and Foxo6 in the injured joints of AB-treated animals. Our results indicate that a reduced state of inflammation at the time of injury and a lower expression of Wnt signaling modulatory protein, Rspo1, caused by AB treatment can slow down or improve PTOA outcomes.
Collapse
Affiliation(s)
- Melanie E. Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Aimy Sebastian
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| | - Summer A. McCloy
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Edward A. Kuhn
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | | | - Gabriela G. Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| |
Collapse
|
32
|
Zhang H, Zhang Y, Terajima M, Romanowicz G, Liu Y, Omi M, Bigelow E, Joiner DM, Waldorff EI, Zhu P, Raghavan M, Lynch M, Kamiya N, Zhang R, Jepsen KJ, Goldstein S, Morris MD, Yamauchi M, Kohn DH, Mishina Y. Loss of BMP signaling mediated by BMPR1A in osteoblasts leads to differential bone phenotypes in mice depending on anatomical location of the bones. Bone 2020; 137:115402. [PMID: 32360900 PMCID: PMC7354232 DOI: 10.1016/j.bone.2020.115402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022]
Abstract
Bone morphogenetic protein (BMP) signaling in osteoblasts plays critical roles in skeletal development and bone homeostasis. Our previous studies showed loss of function of BMPR1A, one of the type 1 receptors for BMPs, in osteoblasts results in increased trabecular bone mass in long bones due to an imbalance between bone formation and bone resorption. Decreased bone resorption was associated with an increased mature-to-immature collagen cross-link ratio and mineral-matrix ratios in the trabecular compartments, and increased tissue-level biomechanical properties. Here, we investigated the bone mass, bone composition and biomechanical properties of ribs and spines in the same genetically altered mouse line to compare outcomes by loss of BMPR1A functions in bones from different anatomic sites and developmental origins. Bone mass was significantly increased in both cortical and trabecular compartments of ribs with minimal to modest changes in compositions. While tissue-levels of biomechanical properties were not changed between control and mutant animals, whole bone levels of biomechanical properties were significantly increased in association with increased bone mass in the mutant ribs. For spines, mutant bones showed increased bone mass in both cortical and trabecular compartments with an increase of mineral content. These results emphasize the differential role of BMP signaling in osteoblasts in bones depending on their anatomical locations, functional loading requirements and developmental origin.
Collapse
Affiliation(s)
- Honghao Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Yanshuai Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Masahiko Terajima
- School of Dentistry, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Genevieve Romanowicz
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Yangjia Liu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA; School of Life Sciences, Tsinghua University, Beijing, China
| | - Maiko Omi
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Erin Bigelow
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Danese M Joiner
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Erik I Waldorff
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Peizhi Zhu
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Mekhala Raghavan
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Michelle Lynch
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Nobuhiro Kamiya
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA; Tenri University, Nara, Japan
| | - Rongqing Zhang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Karl J Jepsen
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Steve Goldstein
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Michael D Morris
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Mitsuo Yamauchi
- School of Dentistry, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - David H Kohn
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW We reviewed recent progress on the role of sclerostin (SOST) and its effects on the immune system in order to summarize the current state of knowledge in osteoimmunology, in regard to hematopoiesis, lymphopoiesis, and inflammation. RECENT FINDINGS Changes in sclerostin levels affect distinct niches within the bone marrow that support hematopoietic stem cells and B cell development. Sclerostin's regulation of adipogenesis could also be important for immune cell maintenance with age. Surprisingly, B cell development in the bone marrow is influenced by Sost produced by mesenchymal stem cells and osteoblasts, but not by osteocytes. Additionally, extramedullary hematopoiesis in the spleen and increased pro-inflammatory cytokine levels in the bone marrow are observed in global Sost-/- mice. In addition to changes in bone marrow density, sclerostin depletion affects B lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect hematopoiesis. It is therefore important to monitor for hematopoietic changes in patients receiving sclerostin-depleting therapies.
Collapse
Affiliation(s)
- Cristine Donham
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA
| | - Jennifer O Manilay
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA.
- Dept. of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, 5200, Lake Road Merced, North, CA, 95343, USA.
| |
Collapse
|
34
|
Abstract
Osteoblasts are specialized mesenchymal cells that synthesize bone matrix and coordinate the mineralization of the skeleton. These cells work in harmony with osteoclasts, which resorb bone, in a continuous cycle that occurs throughout life. The unique function of osteoblasts requires substantial amounts of energy production, particularly during states of new bone formation and remodelling. Over the last 15 years, studies have shown that osteoblasts secrete endocrine factors that integrate the metabolic requirements of bone formation with global energy balance through the regulation of insulin production, feeding behaviour and adipose tissue metabolism. In this article, we summarize the current understanding of three osteoblast-derived metabolic hormones (osteocalcin, lipocalin and sclerostin) and the clinical evidence that suggests the relevance of these pathways in humans, while also discussing the necessity of specific energy substrates (glucose, fatty acids and amino acids) to fuel bone formation and promote osteoblast differentiation.
Collapse
Affiliation(s)
- Naomi Dirckx
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Megan C Moorer
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| |
Collapse
|
35
|
Dzobo K, Thomford NE, Senthebane DA. Targeting the Versatile Wnt/β-Catenin Pathway in Cancer Biology and Therapeutics: From Concept to Actionable Strategy. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:517-538. [PMID: 31613700 DOI: 10.1089/omi.2019.0147] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This expert review offers a critical synthesis of the latest insights and approaches at targeting the Wnt/β-catenin pathway in various cancers such as colorectal cancer, melanoma, leukemia, and breast and lung cancers. Notably, from organogenesis to cancer, the Wnt/β-catenin signaling displays varied and highly versatile biological functions in animals, with virtually all tissues requiring the Wnt/β-catenin signaling in one way or the other. Aberrant expression of the members of the Wnt/β-catenin has been implicated in many pathological conditions, particularly in human cancers. Mutations in the Wnt/β-catenin pathway genes have been noted in diverse cancers. Biochemical and genetic data support the idea that inhibition of Wnt/β-catenin signaling is beneficial in cancer therapeutics. The interaction of this important pathway with other signaling systems is also noteworthy, but remains as an area for further research and discovery. In addition, formation of different complexes by components of the Wnt/β-catenin pathway and the precise roles of these complexes in the cytoplasmic milieu are yet to be fully elucidated. This article highlights the latest medical technologies in imaging, single-cell omics, use of artificial intelligence (e.g., machine learning techniques), genome sequencing, quantum computing, molecular docking, and computational softwares in modeling interactions between molecules and predicting protein-protein and compound-protein interactions pertinent to the biology and therapeutic value of the Wnt/β-catenin signaling pathway. We discuss these emerging technologies in relationship to what is currently needed to move from concept to actionable strategies in translating the Wnt/β-catenin laboratory discoveries to Wnt-targeted cancer therapies and diagnostics in the clinic.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dimakatso A Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
36
|
Tsang KY, Cheah KS. The extended chondrocyte lineage: implications for skeletal homeostasis and disorders. Curr Opin Cell Biol 2019; 61:132-140. [PMID: 31541943 DOI: 10.1016/j.ceb.2019.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 01/14/2023]
Abstract
Endochondral bone formation relies on a finely controlled sequence of chondrocyte proliferation, maturation and hypertrophy that establishes the growth plate which, combined with the deposition of bone upon the cartilage template, mediates longitudinal skeletal growth. Recent lineage studies support a chondrocyte-osteoblast differentiation continuum and the presence of skeletal stem cells within cartilage. The biological significance of the lineage extension and the mechanisms controlling the process are unclear. In this review, we describe recent work on the extended chondrocyte-osteoblast lineage and its contribution to the development, growth and repair of bone and to bone disorders that provides insight into the process and the molecular controls involved. The implications for skeletal homeostasis are discussed.
Collapse
Affiliation(s)
- Kwok Yeung Tsang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kathryn Se Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
37
|
Chicana B, Donham C, Millan AJ, Manilay JO. Wnt Antagonists in Hematopoietic and Immune Cell Fate: Implications for Osteoporosis Therapies. Curr Osteoporos Rep 2019; 17:49-58. [PMID: 30835038 PMCID: PMC6715281 DOI: 10.1007/s11914-019-00503-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW We reviewed the current literature on the roles of the Wnt antagonists sclerostin (Sost) and sclerostin-containing domain protein 1 (Sostdc1) on bone homeostasis, the relationship of the hypoxia-inducible factor (Hif) and von Hippel-Lindau (Vhl) pathways on Sost expression, and how changes in bone induced by depletion of Sost, Sostdc1, and Vhl affect hematopoietic cells. RECENT FINDINGS B cell development is adversely affected in Sost-knockout mice and is more severely affected in Vhl-knockout mice. Inflammation in the Sost-/- bone microenvironment could alter hematopoietic stem cell behavior. Sostdc1-/- mice display defects in natural killer cell development and cytotoxicity. Depletion of Sost and Sostdc1 have effects on immune cell function that warrant investigation in patients receiving Wnt antagonist-depleting therapies for treatment of bone diseases. Additional clinical applications for manipulation of Wnt antagonists include cancer immunotherapies, stem cell transplantation, and directed differentiation to immune lineages.
Collapse
Affiliation(s)
- Betsabel Chicana
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA
| | - Cristine Donham
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA
| | - Alberto J Millan
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA
| | - Jennifer O Manilay
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, USA.
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, 5200 North Lake Road, Merced, CA, 95343, USA.
| |
Collapse
|
38
|
Lou J, Han D, Yu H, Yu G, Jin M, Kim SJ. Cytoprotective Effect of Taurine against Hydrogen Peroxide-Induced Oxidative Stress in UMR-106 Cells through the Wnt/β-Catenin Signaling Pathway. Biomol Ther (Seoul) 2018; 26:584-590. [PMID: 30060293 PMCID: PMC6254645 DOI: 10.4062/biomolther.2018.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis development is closely associated with oxidative stress and reactive oxygen species (ROS). Taurine has potential antioxidant effects, but its role in osteoblasts is not clearly understood. The aim of this study was to determine the protective effects and mechanisms of actions of taurine on hydrogen peroxide (H2O2)-induced oxidative stress in osteoblast cells. UMR-106 cells were treated with taurine prior to H2O2 exposure. After treatment, cell viability, apoptosis, intracellular ROS production, malondialdehyde content, and alkaline phosphate (ALP) activity were measured. We also investigated the protein levels of β-catenin, ERK, CHOP and NF-E2-related factor 2 (Nrf2) along with the mRNA levels of Nrf2 downstream antioxidants. The results showed that pretreatment of taurine could reverse the inhibition of cell viability and suppress the induced apoptosis in a dose-dependent manner: taurine significantly reduced H2O2-induced oxidative damage and expression of CHOP, while it induced protein expression of Nrf2 and β-catenin and activated ERK phosphorylation. DKK1, a Wnt/β-catenin signaling inhibitor, significantly suppressed the taurine-induced Nrf2 signaling pathway and increased CHOP. Activation of ERK signaling mediated by taurine in the presence of H2O2 was significantly inhibited by DKK1. These data demonstrated that taurine protects osteoblast cells against oxidative damage via Wnt/β-catenin-mediated activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Jing Lou
- Department of Immunology, Jinzhou Medical University, Jinzhou 121001, China
| | - Donghe Han
- Department of Neurobiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Huihui Yu
- Department of Immunology, Jinzhou Medical University, Jinzhou 121001, China
| | - Guang Yu
- Department of Immunology, Jinzhou Medical University, Jinzhou 121001, China
| | - Meihua Jin
- Department of Immunology, Jinzhou Medical University, Jinzhou 121001, China.,Key Laboratory of Follicular Development and Reproductive Health of Liaoning, Jinzhou 121001, China
| | - Sung-Jin Kim
- Departments of Pharmacology and Toxicology, Metabolic Diseases Research Laboratory, School of Dentistry, KyungHee University, Seoul 02447, Republic of Korea
| |
Collapse
|