1
|
Wang Y, Zhang W, Yang Y, Qin J, Wang R, Wang S, Fu W, Niu Q, Wang Y, Li C, Li H, Zhou Y, Liu M. Osteopontin deficiency promotes cartilaginous endplate degeneration by enhancing the NF-κB signaling to recruit macrophages and activate the NLRP3 inflammasome. Bone Res 2024; 12:53. [PMID: 39242551 PMCID: PMC11379908 DOI: 10.1038/s41413-024-00355-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 09/09/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is a major cause of discogenic pain, and is attributed to the dysfunction of nucleus pulposus, annulus fibrosus, and cartilaginous endplate (CEP). Osteopontin (OPN), a glycoprotein, is highly expressed in the CEP. However, little is known on how OPN regulates CEP homeostasis and degeneration, contributing to the pathogenesis of IDD. Here, we investigate the roles of OPN in CEP degeneration in a mouse IDD model induced by lumbar spine instability and its impact on the degeneration of endplate chondrocytes (EPCs) under pathological conditions. OPN is mainly expressed in the CEP and decreases with degeneration in mice and human patients with severe IDD. Conditional Spp1 knockout in EPCs of adult mice enhances age-related CEP degeneration and accelerates CEP remodeling during IDD. Mechanistically, OPN deficiency increases CCL2 and CCL5 production in EPCs to recruit macrophages and enhances the activation of NLRP3 inflammasome and NF-κB signaling by facilitating assembly of IRAK1-TRAF6 complex, deteriorating CEP degeneration in a spatiotemporal pattern. More importantly, pharmacological inhibition of the NF-κB/NLRP3 axis attenuates CEP degeneration in OPN-deficient IDD mice. Overall, this study highlights the importance of OPN in maintaining CEP and disc homeostasis, and proposes a promising therapeutic strategy for IDD by targeting the NF-κB/NLRP3 axis.
Collapse
Affiliation(s)
- Yanqiu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wanqian Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Jinghao Qin
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ruoyu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Wenjuan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yanxia Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hongli Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China.
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
2
|
Faggion S, Bonfatti V, Carnier P. Genome-Wide Association Study for Weight Loss at the End of Dry-Curing of Hams Produced from Purebred Heavy Pigs. Animals (Basel) 2024; 14:1983. [PMID: 38998095 PMCID: PMC11240668 DOI: 10.3390/ani14131983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Dissecting the genetics of production traits in livestock is of outmost importance, both to understand biological mechanisms underlying those traits and to facilitate the design of selection programs incorporating that information. For the pig industry, traits related to curing are key for protected designation of origin productions. In particular, appropriate ham weight loss after dry-curing ensures high quality of the final product and avoids economic losses. In this study, we analyzed data (N = 410) of ham weight loss after approximately 20 months of dry-curing. The animals used for ham production were purebred pigs belonging to a commercial line. A genome-wide association study (GWAS) of 29,844 SNP markers revealed the polygenic nature of the trait: 221 loci explaining a small percentage of the variance (0.3-1.65%) were identified on almost all Sus scrofa chromosomes. Post-GWAS analyses revealed 32 windows located within regulatory regions and 94 windows located in intronic regions of specific genes. In total, 30 candidate genes encoding receptors and enzymes associated with ham weight loss (MTHFD1L, DUSP8), proteolysis (SPARCL1, MYH8), drip loss (TNNI2), growth (CDCA3, LSP1, CSMD1, AP2A2, TSPAN4), and fat metabolism (AGPAT4, IGF2R, PTDSS2, HRAS, TALDO1, BRSK2, TNNI2, SYT8, GTF2I, GTF2IRD1, LPCAT3, ATN1, GNB3, CMIP, SORCS2, CCSER1, SPP1) were detected.
Collapse
Affiliation(s)
- Sara Faggion
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Padova, Italy
| | - Valentina Bonfatti
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Padova, Italy
| | - Paolo Carnier
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Padova, Italy
| |
Collapse
|
3
|
Zhou Y, Ni Y, Wang Z, Prud'homme GJ, Wang Q. Causal effects of non-alcoholic fatty liver disease on osteoporosis: a Mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1283739. [PMID: 38149094 PMCID: PMC10749958 DOI: 10.3389/fendo.2023.1283739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Background Osteoporosis (OP) is a systemic skeletal disease characterized by compromised bone strength leading to an increased risk of fracture. There is an ongoing debate on whether non-alcoholic fatty liver disease (NAFLD) is an active contributor or an innocent bystander in the pathogenesis of OP. The aim of this study was to assess the causal association between NAFLD and OP. Methods We performed two-sample Mendelian randomization (MR) analyses to investigate the causal association between genetically predicted NAFLD [i.e., imaging-based liver fat content (LFC), chronically elevated serum alanine aminotransferase (cALT) and biopsy-confirmed NAFLD] and risk of OP. The inverse variant weighted method was performed as main analysis to obtain the causal estimates. Results Imaging-based LFC and biopsy-confirmed NAFLD demonstrated a suggestive causal association with OP ([odds ratio (OR): 1.003, 95% CI: 1.001-1.004, P < 0.001; OR: 1.001, 95% CI: 1.000-1.002, P = 0.031]). The association between cALT and OP showed a similar direction, but was not statistically significant (OR: 1.001, 95% CI: 1.000-1.002, P = 0.079). Repeated analyses after exclusion of genes associated with confounding factors showed consistent results. Sensitivity analysis indicated low heterogeneity, high reliability and low pleiotropy of the causal estimates. Conclusion The two-sample MR analyses suggest a causal association between genetically predicted NAFLD and OP.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Yunzhi Ni
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Zhihong Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Gerald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Doolittle ML, Khosla S, Saul D. Single-Cell Integration of BMD GWAS Results Prioritize Candidate Genes Influencing Age-Related Bone Loss. JBMR Plus 2023; 7:e10795. [PMID: 37808401 PMCID: PMC10556272 DOI: 10.1002/jbm4.10795] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/17/2023] [Accepted: 06/19/2023] [Indexed: 10/10/2023] Open
Abstract
The regulation of bone mineral density (BMD) is highly influenced by genetics and age. Although genome-wide association studies (GWAS) for BMD have uncovered many genes through their proximity to associated variants (variant nearest-neighbor [VNN] genes), the cell-specific mechanisms of each VNN gene remain unclear. This is primarily due to the inability to prioritize these genes by cell type and age-related expression. Using age-related transcriptomics, we found that the expression of many VNN genes was upregulated in the bone and marrow from aged mice. Candidate genes from GWAS were investigated using single-cell RNA-sequencing (scRNA-seq) datasets to enrich for cell-specific expression signatures. VNN candidate genes are highly enriched in osteo-lineage cells, osteocytes, hypertrophic chondrocytes, and Lepr+ mesenchymal stem cells. These data were used to generate a "blueprint" for Cre-loxp mouse line selection for functional validation of candidate genes and further investigation of their role in BMD maintenance throughout aging. In VNN-gene-enriched cells, Sparc, encoding the extracellular matrix (ECM) protein osteonectin, was robustly expressed. This, along with expression of numerous other ECM genes, indicates that many VNN genes likely have roles in ECM deposition by osteoblasts. Overall, we provide data supporting streamlined translation of GWAS candidate genes to potential novel therapeutic targets for the treatment of osteoporosis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Madison L. Doolittle
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Sundeep Khosla
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Dominik Saul
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department for Trauma and Reconstructive SurgeryBG Clinic, University of TuebingenTuebingenGermany
| |
Collapse
|
5
|
Hild V, Mellert K, Möller P, Barth TFE. Giant Cells of Various Lesions Are Characterised by Different Expression Patterns of HLA-Molecules and Molecules Involved in the Cell Cycle, Bone Metabolism, and Lineage Affiliation: An Immunohistochemical Study with a Review of the Literature. Cancers (Basel) 2023; 15:3702. [PMID: 37509363 PMCID: PMC10377796 DOI: 10.3390/cancers15143702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Giant cells (GCs) are thought to originate from the fusion of monocytic lineage cells and arise amid multiple backgrounds. To compare GCs of different origins, we immunohistochemically characterised the GCs of reactive and neoplastic lesions (n = 47). We studied the expression of 15 molecules including HLA class II molecules those relevant to the cell cycle, bone metabolism and lineage affiliation. HLA-DR was detectable in the GCs of sarcoidosis, sarcoid-like lesions, tuberculosis, and foreign body granuloma. Cyclin D1 was expressed by the GCs of neoplastic lesions as well as the GCs of bony callus, fibroid epulis, and brown tumours. While cyclin E was detected in the GCs of all lesions, p16 and p21 showed a heterogeneous expression pattern. RANK was expressed by the GCs of all lesions except sarcoid-like lesions and xanthogranuloma. All GCs were RANK-L-negative, and the GCs of all lesions were osteoprotegerin-positive. Osteonectin was limited to the GCs of chondroblastoma. Osteopontin and TRAP were detected in the GCs of all lesions except xanthogranuloma. RUNX2 was heterogeneously expressed in the reactive and neoplastic cohort. The GCs of all lesions except foreign body granuloma expressed CD68, and all GCs were CD163- and langerin-negative. This profiling points to a functional diversity of GCs despite their similar morphology.
Collapse
Affiliation(s)
- Vivien Hild
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Kevin Mellert
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Peter Möller
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Thomas F E Barth
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
6
|
Osteopontin: A Bone-Derived Protein Involved in Rheumatoid Arthritis and Osteoarthritis Immunopathology. Biomolecules 2023; 13:biom13030502. [PMID: 36979437 PMCID: PMC10046882 DOI: 10.3390/biom13030502] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Osteopontin (OPN) is a bone-derived phosphoglycoprotein related to physiological and pathological mechanisms that nowadays has gained relevance due to its role in the immune system response to chronic degenerative diseases, including rheumatoid arthritis (RA) and osteoarthritis (OA). OPN is an extracellular matrix (ECM) glycoprotein that plays a critical role in bone remodeling. Therefore, it is an effector molecule that promotes joint and cartilage destruction observed in clinical studies, in vitro assays, and animal models of RA and OA. Since OPN undergoes multiple modifications, including posttranslational changes, proteolytic cleavage, and binding to a wide range of receptors, the mechanisms by which it produces its effects, in some cases, remain unclear. Although there is strong evidence that OPN contributes significantly to the immunopathology of RA and OA when considering it as a common denominator molecule, some experimental trial results argue for its protective role in rheumatic diseases. Elucidating in detail OPN involvement in bone and cartilage degeneration is of interest to the field of rheumatology. This review aims to provide evidence of the OPN’s multifaceted role in promoting joint and cartilage destruction and propose it as a common denominator of AR and OA immunopathology.
Collapse
|
7
|
Kim DY, Ko SH. Common Regulators of Lipid Metabolism and Bone Marrow Adiposity in Postmenopausal Women. Pharmaceuticals (Basel) 2023; 16:322. [PMID: 37259464 PMCID: PMC9967016 DOI: 10.3390/ph16020322] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 09/13/2024] Open
Abstract
A variety of metabolic disorders are associated with a decrease in estradiol (E2) during natural or surgical menopause. Postmenopausal women are prone to excessive fat accumulation in skeletal muscle and adipose tissue due to the loss of E2 via abnormalities in lipid metabolism and serum lipid levels. In skeletal muscle and adipose tissue, genes related to energy metabolism and fatty acid oxidation, such as those encoding peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) and estrogen-related receptor alpha (ERRα), are downregulated, leading to increased fat synthesis and lipid metabolite accumulation. The same genes regulate lipid metabolism abnormalities in the bone marrow. In this review, abnormalities in lipid metabolism caused by E2 deficiency were investigated, with a focus on genes able to simultaneously regulate not only skeletal muscle and adipose tissue but also bone metabolism (e.g., genes encoding PGC-1α and ERRα). In addition, the mechanisms through which mesenchymal stem cells lead to adipocyte differentiation in the bone marrow as well as metabolic processes related to bone marrow adiposity, bone loss, and osteoporosis were evaluated, focusing on the loss of E2 and lipid metabolic alterations. The work reviewed here suggests that genes underlying lipid metabolism and bone marrow adiposity are candidate therapeutic targets for bone loss and osteoporosis in postmenopausal women.
Collapse
Affiliation(s)
- Dae-Yong Kim
- CEO, N- BIOTEK, Inc., 402-803, Technopark, 655, Pyeongcheon-ro, Bucheon-si 14502, Gyeonggi-do, Republic of Korea
| | - Seong-Hee Ko
- Regenerative Medicine Research Team, N- BIOTEK, Inc., 104-706, Technopark Ssangyong 3Cha, 397, Seokcheon-ro, Bucheon-si 14449, Gyeonggi-do, Republic of Korea
| |
Collapse
|
8
|
Zhao J, Lei H, Wang T, Xiong X. Liver-bone crosstalk in non-alcoholic fatty liver disease: Clinical implications and underlying pathophysiology. Front Endocrinol (Lausanne) 2023; 14:1161402. [PMID: 36967758 PMCID: PMC10036806 DOI: 10.3389/fendo.2023.1161402] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Osteoporosis is a common complication of many types of chronic liver diseases (CLDs), such as cholestatic liver disease, viral hepatitis, and alcoholic liver disease. Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent metabolic liver disease, affecting almost one third of adults around the world, and is emerging as the dominant cause of CLDs. Liver serves as a hub for nutrient and energy metabolism in the body, and its crosstalk with other tissues, such as adipose tissue, heart, and brain, has been well recognized. However, much less is known about the crosstalk that occurs between the liver and bone. Moreover, the mechanisms by which CLDs increase the risk for osteoporosis remain unclear. This review summarizes the latest research on the liver-bone axis and discusses the relationship between NAFLD and osteoporosis. We cover key signaling molecules secreted by liver, such as insulin-like growth factor-1 (IGF-1), fibroblast growth factor 21 (FGF21), insulin-like growth factor binding protein 1 (IGFBP1), fetuin-A, tumor necrosis factor-alpha (TNF-α), and osteopontin (OPN), and their relevance to the homeostasis of bone metabolism. Finally, we consider the disordered liver metabolism that occurs in patients with NAFLD and how this disrupts signaling to the bone, thereby perturbing the balance between osteoclasts and osteoblasts and leading to osteoporosis or hepatic osteodystrophy (HOD).
Collapse
|
9
|
Deletion of osteopontin or bone sialoprotein induces opposite bone responses to mechanical stimulation in mice. Bone Rep 2022; 17:101621. [PMID: 36159882 PMCID: PMC9493388 DOI: 10.1016/j.bonr.2022.101621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/21/2022] Open
Abstract
Osteopontin (OPN) and Bone Sialoprotein (BSP) are co-expressed in bone and display overlapping and complementary physiological properties. Both genes show a rapid expression response to mechanical stimulation. We used mice with single and double deletions (DKO) of BSP and OPN to assess the specificity of their roles in skeletal adaptation to loading. Two-month-old Wild-Type (WT), BSP knockout (BSP−/−), OPN−/− and DKO male mice were submitted to two mechanical stimulation regimen (n = 10 mice/group) respectively impacting trabecular bone (Hypergravity, HG) and cortical bone (Whole Body Vibration, WBV). HG increased trabecular bone volume (BV/TV) in WT femur through reduced resorption, and in BSP−/− mice femur and vertebra through increased bone formation. In contrast, HG increased the turnover of OPN−/− bone, resulting in reduced femur and vertebra BV/TV. HG did not affect DKO bones. Similarly, WBV increased cortical thickness in BSP−/− mice and decreased it in OPN−/−, without affecting structurally WT and DKO bone. Vibrated BSP−/− mice displayed increased endocortical bone formation with a drop in Sclerostin expression, and reduced periosteal osteoclasts with lower Rankl and Cathepsin K expression. In contrast, vibrated OPN−/− endocortical bone displayed decreased formation and increased osteoclast coverage. Therefore, under two regimen (HG and WBV) targeting distinct bone compartments, absence of OPN resulted in bone loss while lack of BSP induced bone gain, reflecting divergent structural adaptations. Strikingly, absence of both proteins led to a relative insensitivity to either mechanical challenge. Interplay between OPN and BSP thus appears as a key element of skeletal response to mechanical stimulation. Osteopontin gene knockout induces bone loss under mechanical stimulation Bone Sialoprotein gene knockout potentiates bone gain under mechanical stimulation Knockout of both genes leads to bone insensitivity Their interplay is crucial for bone response to mechanical challenges
Collapse
|
10
|
Huang R, Balu AR, Molitoris KH, White JP, Robling AG, Ayturk UM, Baht GS. The role of Meteorin-like in skeletal development and bone fracture healing. J Orthop Res 2022; 40:2510-2521. [PMID: 35076116 PMCID: PMC9309188 DOI: 10.1002/jor.25286] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/11/2022] [Accepted: 01/23/2022] [Indexed: 02/04/2023]
Abstract
Meteorin-like protein (Metrnl), homologous to the initially identified neurotrophic factor Meteorin, is a secreted, multifunctional protein. Here we used mouse models to investigate Metrnl's role in skeletal development and bone fracture healing. During development Metrnl was expressed in the perichondrium and primary ossification center. In neonates, single cell RNA-seq of diaphyseal bone demonstrated strongest expression of Metrnl transcript by osteoblasts. In vitro, Metrnl was osteoinductive, increasing osteoblast differentiation and mineralization in tissue culture models. In vivo, loss of Metrnl expression resulted in no change in skeletal metrics in utero, at birth, or during postnatal growth. Six-week-old Metrnl-null mice displayed similar body length, body weight, tibial length, femoral length, BV/TV, trabecular number, trabecular thickness, and cortical thickness as littermate controls. In 4-month-old mice, lack of Metrnl expression did not change structural stiffness, ultimate force, or energy to fracture of femora under 3-point-bending. Last, we investigated the role of Metrnl in bone fracture healing. Metrnl expression increased in response to tibial injury, however, loss of Metrnl expression did not affect the amount of bone deposited within the healing tissue nor did it change the structural parameters of healing tissue. This work identifies Metrnl as a dispensable molecule for skeletal development. However, the osteoinductive capabilities of Metrnl may be utilized to modulate osteoblast differentiation in cell-based orthopedic therapies.
Collapse
Affiliation(s)
- Rong Huang
- Department of MedicineDuke Molecular Physiology InstituteDurhamNorth CarolinaUSA,Department of Orthopaedic SurgeryDuke UniversityDurhamNorth CarolinaUSA
| | - Abhinav R. Balu
- Department of MedicineDuke Molecular Physiology InstituteDurhamNorth CarolinaUSA,Department of Orthopaedic SurgeryDuke UniversityDurhamNorth CarolinaUSA
| | - Kristin H. Molitoris
- Department of MedicineDuke Molecular Physiology InstituteDurhamNorth CarolinaUSA,Department of Orthopaedic SurgeryDuke UniversityDurhamNorth CarolinaUSA
| | - James P. White
- Department of MedicineDuke Molecular Physiology InstituteDurhamNorth CarolinaUSA
| | - Alexander G. Robling
- Department of Anatomy and Cell BiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ugur M. Ayturk
- Department of ResearchHospital for Special SurgeryNew York CityNew YorkUSA,Department of Orthopaedic SurgeryWeill Cornell MedicineNew York CityNew YorkUSA
| | - Gurpreet S. Baht
- Department of MedicineDuke Molecular Physiology InstituteDurhamNorth CarolinaUSA,Department of Orthopaedic SurgeryDuke UniversityDurhamNorth CarolinaUSA,Department of PathologyDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
11
|
Vachliotis ID, Anastasilakis AD, Goulas A, Goulis DG, Polyzos SA. Nonalcoholic fatty liver disease and osteoporosis: A potential association with therapeutic implications. Diabetes Obes Metab 2022; 24:1702-1720. [PMID: 35589613 DOI: 10.1111/dom.14774] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/01/2022] [Accepted: 05/17/2022] [Indexed: 11/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and osteoporosis are two highly prevalent metabolic diseases. Increasing experimental evidence supports a pathophysiological link between NAFLD and osteoporosis. A key feature could be chronic, low-grade inflammation, which characterizes NAFLD and possibly affects bone metabolism. In this context, several factors, including but not limited to receptor activator of nuclear factor kappa-B ligand, osteoprotegerin, osteopontin and osteocalcin, may serve as mediators. In the clinical setting, most but not all epidemiological evidence indicates that NAFLD is associated with lower bone mineral density or osteoporosis in adults. Although an association between NAFLD and osteoporosis has not yet been established, and thus remains speculative, pharmacological considerations already exist. Some of the current and emerging pharmacological options for NAFLD have shown possible anti-osteoporotic properties (eg, vitamin E, obeticholic acid, semaglutide), while others (eg, pioglitazone, canagliflozin) have been associated with increased risk of fractures and may be avoided in patients with NAFLD and concomitant osteoporosis, especially those at high fracture risk. Conversely, some anti-osteoporotic medications (denosumab) might benefit NAFLD, while others (raloxifene) might adversely affect it and, consequently, may be avoided in patients with osteoporosis and NAFLD. If an association between NAFLD and osteoporosis is established, a medication that could target both diseases would be a great advancement. This review summarizes the main experimental and clinical evidence on the potential association between NAFLD and osteoporosis and focuses on treatment considerations derived from this potential association.
Collapse
Affiliation(s)
- Ilias D Vachliotis
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Endocrinology, 424 General Military Hospital, Thessaloniki, Greece
| | | | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
12
|
Feng C, Xu Z, Tang X, Cao H, Zhang G, Tan J. Estrogen-Related Receptor α: A Significant Regulator and Promising Target in Bone Homeostasis and Bone Metastasis. Molecules 2022; 27:3976. [PMID: 35807221 PMCID: PMC9268386 DOI: 10.3390/molecules27133976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/11/2022] [Accepted: 06/20/2022] [Indexed: 01/23/2023] Open
Abstract
Bone homeostasis is maintained with the balance between bone formation and bone resorption, which is involved in the functional performance of osteoblast and osteoclast. Disruption of this equilibrium usually causes bone disorders including osteoporosis, osteoarthritis, and osteosclerosis. In addition, aberrant activity of bone also contributes to the bone metastasis that frequently occurs in the late stage of aggressive cancers. Orphan nuclear receptor estrogen-related receptor (ERRα) has been demonstrated to control the bone cell fate and the progression of tumor cells in bone through crosstalk with various molecules and signaling pathways. However, the defined function of this receptor in bone is inconsistent and controversial. Therefore, we summarized the latest research and conducted an overview to reveal the regulatory effect of ERRα on bone homeostasis and bone metastasis, this review may broaden the present understanding of the cellular and molecular model of ERRα and highlight its potential implication in clinical therapy.
Collapse
Affiliation(s)
- Chun Feng
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, China; (C.F.); (Z.X.)
| | - Zhaowei Xu
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, China; (C.F.); (Z.X.)
| | - Xiaojie Tang
- Department of Spinal Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, China; (X.T.); (H.C.)
| | - Haifei Cao
- Department of Spinal Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, China; (X.T.); (H.C.)
| | - Guilong Zhang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai 264003, China; (C.F.); (Z.X.)
| | - Jiangwei Tan
- Department of Spinal Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, China; (X.T.); (H.C.)
| |
Collapse
|
13
|
Bartlett CL, Cave EM, Crowther NJ, Ferris WF. A new perspective on the function of Tissue Non-Specific Alkaline Phosphatase: from bone mineralization to intra-cellular lipid accumulation. Mol Cell Biochem 2022; 477:2093-2106. [PMID: 35471716 DOI: 10.1007/s11010-022-04429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/31/2022] [Indexed: 11/29/2022]
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is one of four isozymes, which include germ cell, placental and intestinal alkaline phosphatases. The TNAP isozyme has 3 isoforms (liver, bone and kidney) which differ by tissue expression and glycosylation pattern. Despite a long history of investigation, the exact function of TNAP in many tissues is largely unknown. Only the bone isoform has been well characterised during mineralization where the enzyme hydrolyses pyrophosphate to inorganic phosphate, which combines with calcium to form hydroxyapatite crystals deposited as new bone. The inorganic phosphate also increases gene expression of proteins that support tissue mineralization. Recent studies have shown that TNAP is expressed in preadipocytes from several species, and that inhibition of TNAP activity causes attenuation of intracellular lipid accumulation in these and other lipid-storing cells. The mechanism by which TNAP stimulates lipid accumulation is not known; however, proteins that are important for controlling phosphate levels in bone are also expressed in adipocytes. This review examines the evidence that inorganic phosphate generated by TNAP promotes transcription that enhances the expression of the regulators of lipid storage and consequently, that TNAP has a major function of lipid metabolism.
Collapse
Affiliation(s)
- Cara-Lesley Bartlett
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eleanor Margaret Cave
- Department of Chemical Pathology, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Nigel John Crowther
- Department of Chemical Pathology, University of the Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa.,Department of Chemical Pathology, National Health Laboratory Service, Johannesburg, South Africa
| | - William Frank Ferris
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
14
|
Kawtharany L, Bessueille L, Issa H, Hamade E, Zibara K, Magne D. Inflammation and Microcalcification: A Never-Ending Vicious Cycle in Atherosclerosis? J Vasc Res 2022; 59:137-150. [PMID: 35038712 DOI: 10.1159/000521161] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/17/2021] [Indexed: 11/19/2022] Open
Abstract
Inflammatory cells and cytokines are known for long to worsen the development of atherosclerotic plaques in mice, and intense efforts are today devoted to develop anti-inflammatory therapeutic strategies to slow down plaque development. Increasing data indicate that plaque inflammation is intimately associated with microcalcifications, which exert harmful effects eventually culminating with plaque rupture. In this review article, we will first introduce microcalcification location, detection, and effects in atherosclerotic plaques. Then, we will present the numerous data suggesting that inflammatory cells and molecules are responsible for the formation of microcalcifications and the articles showing that microcalcifications stimulate macrophages and smooth muscle cells to produce more pro-inflammatory cytokines. Finally, we will discuss the possibility that microcalcifications might stimulate smooth muscle cells to produce larger and more stable calcifications to stabilize plaques, to exit the vicious cycle associating inflammation and microcalcification in atherosclerotic plaques.
Collapse
Affiliation(s)
- Lynn Kawtharany
- ICBMS UMR CNRS 5246, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Hawraa Issa
- College of Public Health, Phoenicia University, Zahrani, Lebanon
| | - Eva Hamade
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - David Magne
- ICBMS UMR CNRS 5246, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
15
|
Goettsch C, Strzelecka-Kiliszek A, Bessueille L, Quillard T, Mechtouff L, Pikula S, Canet-Soulas E, Luis MJ, Fonta C, Magne D. TNAP as a therapeutic target for cardiovascular calcification: a discussion of its pleiotropic functions in the body. Cardiovasc Res 2022; 118:84-96. [PMID: 33070177 PMCID: PMC8752354 DOI: 10.1093/cvr/cvaa299] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular calcification (CVC) is associated with increased morbidity and mortality. It develops in several diseases and locations, such as in the tunica intima in atherosclerosis plaques, in the tunica media in type 2 diabetes and chronic kidney disease, and in aortic valves. In spite of the wide occurrence of CVC and its detrimental effects on cardiovascular diseases (CVD), no treatment is yet available. Most of CVC involve mechanisms similar to those occurring during endochondral and/or intramembranous ossification. Logically, since tissue-nonspecific alkaline phosphatase (TNAP) is the key-enzyme responsible for skeletal/dental mineralization, it is a promising target to limit CVC. Tools have recently been developed to inhibit its activity and preclinical studies conducted in animal models of vascular calcification already provided promising results. Nevertheless, as its name indicates, TNAP is ubiquitous and recent data indicate that it dephosphorylates different substrates in vivo to participate in other important physiological functions besides mineralization. For instance, TNAP is involved in the metabolism of pyridoxal phosphate and the production of neurotransmitters. TNAP has also been described as an anti-inflammatory enzyme able to dephosphorylate adenosine nucleotides and lipopolysaccharide. A better understanding of the full spectrum of TNAP's functions is needed to better characterize the effects of TNAP inhibition in diseases associated with CVC. In this review, after a brief description of the different types of CVC, we describe the newly uncovered additional functions of TNAP and discuss the expected consequences of its systemic inhibition in vivo.
Collapse
Affiliation(s)
- Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen
University, Aachen, Germany
| | - Agnieszka Strzelecka-Kiliszek
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Laurence Bessueille
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| | - Thibaut Quillard
- PHY-OS Laboratory, UMR 1238 INSERM, Université de Nantes, CHU
de Nantes, France
| | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France
- CREATIS Laboratory, CNRS UMR 5220, Inserm U1044, Université Claude Bernard
Lyon 1, Lyon, France
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, Univ Lyon, INSERM, INRA, INSA Lyon, Université Claude
Bernard Lyon 1, Lyon, France
| | - Millan Jose Luis
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery
Institute, La Jolla, CA 92037, USA
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de
Toulouse, France
| | - David Magne
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| |
Collapse
|
16
|
Bai RJ, Li YS, Zhang FJ. Osteopontin, a bridge links osteoarthritis and osteoporosis. Front Endocrinol (Lausanne) 2022; 13:1012508. [PMID: 36387862 PMCID: PMC9649917 DOI: 10.3389/fendo.2022.1012508] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease characterized by degradation of articular cartilage, inflammation, and changes in periarticular and subchondral bone of joints. Osteoporosis (OP) is another systemic skeletal disease characterized by low bone mass and bone mineral density (BMD) accompanied by microarchitectural deterioration in bone tissue and increased bone fragility and fracture risk. Both OA and OP are mainly affected on the elderly people. Recent studies have shown that osteopontin (OPN) plays a vital role in bone metabolism and homeostasis. OPN involves these biological activities through participating in the proliferation, migration, differentiation, and adhesion of several bone-related cells, including chondrocytes, synoviocytes, osteoclasts, osteoblasts, and marrow mesenchymal stem cells (MSCs). OPN has been demonstrated to be closely related to the occurrence and development of many bone-related diseases, such as OA and OP. This review summarizes the role of OPN in regulating inflammation activity and bone metabolism in OA and OP. Furthermore, some drugs that targeted OPN to treat OA and OP are also summarized in the review. However, the complex mechanism of OPN in regulating OA and OP is not fully elucidated, which drives us to explore the depth effect of OPN on these two bone diseases.
Collapse
Affiliation(s)
- Rui-Jun Bai
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Sheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- *Correspondence: Yu-Sheng Li, ; Fang-Jie Zhang,
| | - Fang-Jie Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Yu-Sheng Li, ; Fang-Jie Zhang,
| |
Collapse
|
17
|
Zuo H, Yang D, Wan Y. Fam20C Regulates Bone Resorption and Breast Cancer Bone Metastasis through Osteopontin and BMP4. Cancer Res 2021; 81:5242-5254. [PMID: 34433585 DOI: 10.1158/0008-5472.can-20-3328] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/14/2020] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
Fam20C is a kinase that generates the majority of secreted phosphoproteins and regulates biomineralization. However, its potential roles in bone resorption and breast cancer bone metastasis are unknown. Here we show that Fam20C in the myeloid lineage suppresses osteoclastogenesis and bone resorption, during which, osteopontin (OPN) is the most abundant phosphoprotein secreted in a Fam20C-dependent manner. OPN phosphorylation by Fam20C decreased OPN secretion, and OPN neutralization reduced Fam20C deficiency-induced osteoclast differentiation and bone metastasis. In contrast, Fam20C in breast cancer cells promoted bone metastasis by facilitating the phosphorylation and secretion of BMP4, which in turn enhanced osteoclastogenesis. Mutation of the BMP4 phosphorylation site elevated BMP4 lysosomal degradation and reduced BMP4 secretion. In breast cancer cells, BMP4 depletion or treatment with a BMP4 signaling inhibitor diminished osteoclast differentiation and bone metastasis and abolished Fam20C-mediated regulation of these processes. Collectively, this study discovers distinct roles for Fam20C in myeloid cells and breast cancer cells and highlights OPN and BMP4 as potential therapeutic targets for breast cancer bone metastasis. SIGNIFICANCE: Osteoclastogenesis and bone metastasis are suppressed by myeloid-derived Fam20C, but enhanced by breast cancer-associated Fam20C, uncovering novel Fam20C functions and new therapeutic strategies via targeting Fam20C substrates OPN and BMP4.
Collapse
Affiliation(s)
- Hao Zuo
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Dengbao Yang
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yihong Wan
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
18
|
Vancea A, Serban O, Fodor D. Relationship between Osteopontin and Bone Mineral Density. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2021; 17:509-516. [PMID: 35747863 DOI: 10.4183/aeb.2021.509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent studies suggest that osteopontin (OPN) could be used as an early marker for the diagnosis of bone disorders. Considering the contradictory opinions in the literature, the objective of this systematic review is to analyse the current information regarding the relationship between OPN and bone mineral density (BMD), which represents an important process in the development of osteoporosis. We performed a literature search of clinical trials using the PubMed database, published between 1999-2020, and identified 7 studies that were eligible for analysis. The eligibility criteria were based on studies that analysed the relationship between osteopontin and bone mineral density on human subjects. Conclusion: serum OPN levels might be used as a biomarker of the early diagnosis of osteoporosis in postmenopausal women, with or without osteoporotic vertebral fractures.
Collapse
Affiliation(s)
- A Vancea
- "Iuliu Hațieganu" University of Medicine and Pharmacy, 2 Internal Medicine Department, Cluj-Napoca, Romania
| | - O Serban
- "Iuliu Hațieganu" University of Medicine and Pharmacy, 2 Internal Medicine Department, Cluj-Napoca, Romania
| | - D Fodor
- "Iuliu Hațieganu" University of Medicine and Pharmacy, 2 Internal Medicine Department, Cluj-Napoca, Romania
| |
Collapse
|
19
|
Chen H, Shang D, Wen Y, Liang C. Bone-Derived Modulators That Regulate Brain Function: Emerging Therapeutic Targets for Neurological Disorders. Front Cell Dev Biol 2021; 9:683457. [PMID: 34179014 PMCID: PMC8222721 DOI: 10.3389/fcell.2021.683457] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
Bone has traditionally been regarded as a structural organ that supports and protects the various organs of the body. Recent studies suggest that bone also acts as an endocrine organ to regulate whole-body metabolism. Particularly, homeostasis of the bone is shown to be necessary for brain development and function. Abnormal bone metabolism is associated with the onset and progression of neurological disorders. Recently, multiple bone-derived modulators have been shown to participate in brain function and neurological disorders, including osteocalcin, lipocalin 2, and osteopontin, as have bone marrow-derived cells such as mesenchymal stem cells, hematopoietic stem cells, and microglia-like cells. This review summarizes current findings regarding the roles of these bone-derived modulators in the brain, and also follows their involvement in the pathogenesis of neurological disorders. The content of this review may aide in the development of promising therapeutic strategies for neurological disorders via targeting bone.
Collapse
Affiliation(s)
- Hongzhen Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China.,Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuguan Wen
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
20
|
Salivary Osteopontin as a Potential Biomarker for Oral Mucositis. Metabolites 2021; 11:metabo11040208. [PMID: 33808230 PMCID: PMC8066152 DOI: 10.3390/metabo11040208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN), a multifunctional phosphoglycoprotein also presents in saliva, plays a crucial role in tumour progression, inflammation and mucosal protection. Mucosal barrier injury due to high-dose conditioning regimen administered during autologous and allogeneic peripheral stem cell transplantation (APSCT) has neither efficient therapy nor established biomarkers. Our aim was to assess the biomarker role of OPN during APSCT, with primary focus on oral mucositis (OM). Serum and salivary OPN levels were determined by ELISA in 10 patients during APSCT at four stages of transplantation (day -3/-7, 0, +7, +14), and in 23 respective healthy controls. Results: There was a negative correlation between both salivary and serum OPN levels and grade of OM severity during APSCT (r = -0.791, p = 0.019; r = -0.973, p = 0.001). Salivary OPN increased at days +7 (p = 0.011) and +14 (p = 0.034) compared to controls. Among patients, it was higher at day +14 compared to the time of admission (day -3/-7) (p = 0.039) and transplantation (day 0) (p = 0.011). Serum OPN remained elevated at all four stages of transplantation compared to controls (p = 0.013, p = 0.02, p = 0.011, p = 0.028). During APSCT elevated salivary OPN is a potential non-invasive biomarker of oral mucositis whereas the importance of high serum OPN warrants further studies.
Collapse
|
21
|
Depalle B, McGilvery CM, Nobakhti S, Aldegaither N, Shefelbine SJ, Porter AE. Osteopontin regulates type I collagen fibril formation in bone tissue. Acta Biomater 2021; 120:194-202. [PMID: 32344173 PMCID: PMC7821990 DOI: 10.1016/j.actbio.2020.04.040] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/31/2020] [Accepted: 04/21/2020] [Indexed: 01/06/2023]
Abstract
Osteopontin (OPN) is a non-collagenous protein involved in biomineralization of bone tissue. Beyond its role in biomineralization, we show that osteopontin is essential to the quality of collagen fibrils in bone. Transmission electron microscopy revealed that, in Opn-/- tissue, the organization of the collagen fibrils was highly heterogeneous, more disorganized than WT bone and comprised of regions of both organized and disorganized matrix with a reduced density. The Opn-/- bone tissue also exhibited regions in which the collagen had lost its characteristic fibrillar structure, and the crystals were disorganized. Using nanobeam electron diffraction, we show that damage to structural integrity of collagen fibrils in Opn-/- bone tissue and their organization causes mineral disorganization, which could ultimately affect its mechanical integrity. STATEMENT OF SIGNIFICANCE: This study presents new evidence about the role of osteopontin (OPN) - a non-collagenous protein - on the structure and organization of the organic and mineral matrix in bone. In previous work, osteopontin has been suggested to regulate the nucleation and growth of bone mineral crystals and to form sacrificial bonds between mineralized collagen fibrils to enhance bone's toughness. Our findings show that OPN plays a crucial role before mineralization, during the formation of the collagen fibrils. OPN-deficient bones present a lower collagen content compared to wild type bone and, at the tissue level, collagen fibrils organization can be significantly altered in the absence of OPN. Our results suggest that OPN is critical for the formation and/or remodeling of bone collagen matrix. Our findings could lead to the development of new therapeutic strategies of bone diseases affecting collagen formation and remodeling.
Collapse
Affiliation(s)
- Baptiste Depalle
- Department of Materials Science and Engineering, Imperial College London, London, United Kingdom; The Forsyth Institute, Cambridge, MA United States.
| | - Catriona M McGilvery
- Department of Materials Science and Engineering, Imperial College London, London, United Kingdom
| | - Sabah Nobakhti
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA United States
| | - Nouf Aldegaither
- Department of Materials Science and Engineering, Imperial College London, London, United Kingdom; College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Sandra J Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA United States; Department of Bioengineering, Northeastern University, Boston, MA United States
| | - Alexandra E Porter
- Department of Materials Science and Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
22
|
Bailey S, Sroga GE, Hoac B, Katsamenis OL, Wang Z, Bouropoulos N, McKee MD, Sørensen ES, Thurner PJ, Vashishth D. The role of extracellular matrix phosphorylation on energy dissipation in bone. eLife 2020; 9:58184. [PMID: 33295868 PMCID: PMC7746230 DOI: 10.7554/elife.58184] [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: 04/23/2020] [Accepted: 12/07/2020] [Indexed: 01/22/2023] Open
Abstract
Protein phosphorylation, critical for cellular regulatory mechanisms, is implicated in various diseases. However, it remains unknown whether heterogeneity in phosphorylation of key structural proteins alters tissue integrity and organ function. Here, osteopontin phosphorylation level declined in hypo- and hyper- phosphatemia mouse models exhibiting skeletal deformities. Phosphorylation increased cohesion between osteopontin polymers, and adhesion of osteopontin to hydroxyapatite, enhancing energy dissipation. Fracture toughness, a measure of bone’s mechanical competence, increased with ex-vivo phosphorylation of wildtype mouse bones and declined with ex-vivo dephosphorylation. In osteopontin-deficient mice, global matrix phosphorylation level was not associated with toughness. Our findings suggest that phosphorylated osteopontin promotes fracture toughness in a dose-dependent manner through increased interfacial bond formation. In the absence of osteopontin, phosphorylation increases electrostatic repulsion, and likely protein alignment and interfilament distance leading to decreased fracture resistance. These mechanisms may be of importance in other connective tissues, and the key to unraveling cell–matrix interactions in diseases.
Collapse
Affiliation(s)
- Stacyann Bailey
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, United States
| | - Grazyna E Sroga
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, United States
| | - Betty Hoac
- Faculty of Dentistry, McGill University, Montreal, Canada
| | - Orestis L Katsamenis
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom
| | - Zehai Wang
- Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, United States
| | | | - Marc D McKee
- Faculty of Dentistry, McGill University, Montreal, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Canada
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Philipp J Thurner
- Institute of Lightweight Design and Structural Biomechanics, Vienna University of Technology, Vienna, Austria
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, United States
| |
Collapse
|
23
|
Simfia I, Schiavi J, McNamara LM. ROCK-II inhibition suppresses impaired mechanobiological responses in early estrogen deficient osteoblasts. Exp Cell Res 2020; 396:112264. [PMID: 32898551 DOI: 10.1016/j.yexcr.2020.112264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/22/2022]
Abstract
Mechanobiological responses by osteoblasts are governed by downstream Rho-ROCK signalling through actin cytoskeleton re-arrangements but whether these responses are influenced by estrogen deficiency during osteoporosis remains unknown. The objective of this study was to determine alterations in the mechanobiological responses of estrogen-deficient osteoblasts and investigate whether an inhibitor of the Rho-ROCK signalling can revert these changes. MC3T3-E1 cells were pre-treated with 10 nM 17-β estradiol for 7 days and further cultured with or without estradiol for next 2 days. These cells were treated with or without ROCK-II inhibitor, Y-27632, and oscillatory fluid flow (OFF, 1Pa, 0.5 Hz, 1 h) was applied. Here, we report that Prostaglandin E2 release, Runt-related transcription factor 2 and Osteopontin gene expression were significantly enhanced in response to OFF in estrogen-deficient cells than in cells with estrogen (3.73 vs 1.63 pg/ng DNA; 13.5 vs 2.6 fold, 2.1 vs 0.4 fold respectively). Upon ROCK-II inhibition, these enhanced effects of estrogen deficiency were downregulated. OFF increased the fibril anisotropy in cells pre-treated with estrogen and this increase was suppressed upon ROCK-II inhibition. This study is the first to demonstrate altered mechanobiological responses by osteoblasts during early estrogen deficiency and that these responses to OFF can be suppressed upon ROCK inhibition.
Collapse
Affiliation(s)
- Irene Simfia
- Mechanobiology and Medical Device Research Group, Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Jessica Schiavi
- Mechanobiology and Medical Device Research Group, Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Laoise M McNamara
- Mechanobiology and Medical Device Research Group, Biomechanics Research Centre, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
24
|
Miki H, Okito A, Akiyama M, Ono T, Tachikawa N, Nakahama KI. Genetic and epigenetic regulation of osteopontin by cyclic adenosine 3' 5'-monophosphate in osteoblasts. Gene 2020; 763:145059. [PMID: 32858177 DOI: 10.1016/j.gene.2020.145059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/20/2020] [Accepted: 08/12/2020] [Indexed: 01/02/2023]
Abstract
Osteopontin (OPN) is not only a marker of osteoblasts but it is also related to cancer progression and inflammation. The expression of OPN increases in response to inflammatory cytokines, hormones, and mechanical stress. Among them, cyclic-AMP (cAMP) elevating agents stimulate OPN expression in the presence of 1, 25-OH vitamin D3 (VD3). We aimed to clarify the mechanism by which cAMP enhances OPN expression in osteoblastic cells. The OPN promoter (-2335 to +76, OPNp2335) exerted a cell type specific response to forskolin (FK) and VD3. Sequential deletion analysis of OPNp revealed that the OPNp (-833 to +76) contained essential responsive regions to respond to cAMP signaling. In particular, both Vitamin D response element (VDRE, -758 to -743) and osteoblast-specific cis- acting element 2 (OSE2, -695 to -690) were essential for cAMP-mediated OPNp activity. The expression of vitamin D receptor (VDR), but not runt-related transcription factor 2 (Runx2), a nuclear receptor for OSE2, was induced by the treatment of the cells with FK. Although, VD3-induced OPNp activity was slightly enhanced in VDR-overexpressing osteoblasts, it reached the same level as that of osteoblasts induced by both VD3 and FK in the presence of histone deacetylase (HDAC) inhibitor. Moreover, we identified histone acetylation on the OPN promoter region by FK treatment. These results strongly suggest that OPNp activity is controlled by the cAMP signaling via genetic and epigenetic regulations.
Collapse
Affiliation(s)
- Hirohito Miki
- Department of Cellular Physiological Chemistry, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan
| | - Asuka Okito
- Department of Cellular Physiological Chemistry, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Department of Orthodontic Science, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masako Akiyama
- Research Administration Division, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Noriko Tachikawa
- Oral Implantology and Regenerative Dental Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan
| | - Ken-Ichi Nakahama
- Department of Cellular Physiological Chemistry, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
25
|
Wu LF, Wang WY, Zhu DC, He P, Zhu K, Gui GP, Gao HQ, Mo XB, Lu X, Deng FY, Lei SF. Protein array test detected three osteoporosis related plasma inflammatory cytokines in Chinese postmenopausal women. Cytokine 2020; 133:155166. [PMID: 32570159 DOI: 10.1016/j.cyto.2020.155166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023]
Abstract
Inflammatory cytokines were involved in pathological conditions of osteoporosis (OP). However, the specific OP-associated inflammatory cytokines are still awaiting to be detected by using a systemic method. Herein, we adopted an extreme sampling scheme and examined inflammatory cytokines between subjects with low and high bone mineral density (BMD) through protein microarray. First, 8 candidate cytokines including B lymphocyte chemoattractant (BLC), osteopontin (OPN) and insulin-like growth factor-binding protein 4 (IGFBP4) were identified in the discovery extreme sampling subgroup. Then, the different expressions for BLC, OPN and IGFBP4 were validated and replicated in two independent extreme sampling subgroups. Further functional experiments showed that the cytokine BLC was involved in bone metabolism by inhibiting bone formation and promoting bone resorption. Together, this study further revealed that inflammatory cytokines were closely related with OP, and that they highlighted critical roles of BLC in the pathogenesis of OP.
Collapse
Affiliation(s)
- Long-Fei Wu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wen-Yu Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China; Suzhou Center for Disease Prevention and Control, Suzhou, Jiangsu, China
| | - Dong-Cheng Zhu
- Department of Orthopedics, Sihong People's Hospital, Suqian, Jiangsu 223900, China
| | - Pei He
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kan Zhu
- Loujiang Community Health Service Center, Suzhou Gusu District, Suzhou, Jiangsu, China
| | - Guo-Ping Gui
- Disease Prevention and Control Center of Suzhou High Tech Zone, Suzhou, Jiangsu, China
| | - Hong-Qin Gao
- Shishan Community Health Service Center, Suzhou High Tech Zone, Suzhou, Jiangsu, China
| | - Xing-Bo Mo
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xin Lu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fei-Yan Deng
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
26
|
Shaalan AAM, El-Sherbiny M, El-Abaseri TB, Shoaeir MZ, Abdel-Aziz TM, Mohamed MI, Zaitone SA, Mohammad HMF. Supplement With Calcium or Alendronate Suppresses Osteopenia Due to Long Term Rabeprazole Treatment in Female Mice: Influence on Bone TRAP and Osteopontin Levels. Front Pharmacol 2020; 11:583. [PMID: 32477111 PMCID: PMC7237708 DOI: 10.3389/fphar.2020.00583] [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: 02/16/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background and Purpose Rabeprazole, a proton pump inhibitor (PPIs) is much endorsed to patients with increased gastric acidity. PPIs were accused to have osteoporotic effects on patients who chronically use them. The point of the current investigation was to decide the impact of rabeprazole on osteoporosis and to explore the modulatory effects of dietary calcium or alendronate on this side effect. Methods 80 female mice were alienated into four groups maintained for 18 weeks: [1] Vehicle group: given distilled water in 12 ml/kg, P.O. [2] Rabeprazole control group: given rabeprazole in a dose equals 10 mg/kg every 48 h, P.O. [3] Rabeprazole + calcium: given rabeprazole (10 mg/kg every 48 h) along with calcium supplement. [4] Rabeprazole + alendronate: given rabeprazole (10 mg/kg every 48 h) and alendronate (1 mg/kg per week, i.p.). Serum calcium, phosphorus and parathyroid hormone were measured. Both femurs were kept in paraformaldehyde, and then the right one was used for X-ray examination with analysis by Digora software and the left one for histopathological examination (H&E) and immunohistochemical stains for osteopontin and tartrate resistant acid phosphatase (TRAP). Results Calcium supplementation or administration of alendronate along with rabeprazole significantly restored the mean bone density as shown by X-ray analysis. Femurs from mice received rabeprazole showed widely separated, thin-walled bone trabeculae and increased number of osteoclasts. Calcium or alendronate with rabeprazole showed thick bone trabeculae without full recovery from rabeprazole induced damage. Adding calcium supplementation to rabeprazole did not affect the histological abnormalities related to osteoclasts meanwhile alendronate produced inactivation of osteoclasts. Both calcium and alendronate decreased the rabeprazole-induced increment in the femur osteopontin level. Conclusion Calcium or alendronate can be recommended for female patients on PPI therapy who are at risk of osteopenia.
Collapse
Affiliation(s)
- Aly A M Shaalan
- Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,Department of Anatomy, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Department of Anatomy, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Taghrid B El-Abaseri
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Mohamed Z Shoaeir
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Tarek M Abdel-Aziz
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Magda I Mohamed
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Hala M F Mohammad
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,Central Laboratory, Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
27
|
Cleverdon RE, McAlpine MD, Ward WE. Black Tea Exhibits a Dose-Dependent Response in Saos-2 Cell Mineralization. J Med Food 2020; 23:1014-1018. [PMID: 32208048 PMCID: PMC7478194 DOI: 10.1089/jmf.2020.0010] [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] [Indexed: 01/05/2023] Open
Abstract
Higher bone mineral density (BMD) is often associated with greater consumption of black tea (BT). However, the dose–response of BT on mineralization in an osteoblast cell model has not yet been studied. The study objective was to determine the dose-dependent response of BT in Saos-2 cells and investigate changes to several proteins involved in the mineralization process. Mineralization was induced in the presence of BT at concentrations that represent levels likely achieved through daily consumption (0.1, 0.5, 0.75, 1 μg gallic acid equivalents [GAE]/mL) or through supplementation (2, 5, or 10 μg GAE/mL). BT exerted a positive dose–response on bone mineralization, peaking at 1 μg GAE/mL of BT (P < .05). Cellular activity was significantly greater than control with exposure to 2–10 μg GAE/mL of BT (at 24 h) (P < .05) and 1–10 μg GAE/mL (at 48 h) (P < .05), with a peak at 5 μg GAE/mL at 24 and 48 h (P < .05). Protein expression of alkaline phosphatase and ectonucleotide pyrophosphatase/phosphodiesterase-1 were unchanged, whereas a moderate dose of BT (0.75 μg GAE/mL) resulted in greater expression of osteopontin compared with the highest dose (10 μg GAE/mL) (P < .05). Doses of BT from 0.5 to 10 μg GAE/mL resulted in higher antioxidant capacity compared with control (P < .05). In summary, the higher antioxidant capacity, enhanced cell viability, and upregulated mineralization suggest that consumption of BT may have a positive effect on BMD at levels obtained through consumption of tea.
Collapse
Affiliation(s)
- Riley E Cleverdon
- Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Muscle and Bone Health, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Michael D McAlpine
- Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Muscle and Bone Health, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Wendy E Ward
- Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada.,Centre for Muscle and Bone Health, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
28
|
Ragipoglu D, Dudeck A, Haffner-Luntzer M, Voss M, Kroner J, Ignatius A, Fischer V. The Role of Mast Cells in Bone Metabolism and Bone Disorders. Front Immunol 2020; 11:163. [PMID: 32117297 PMCID: PMC7025484 DOI: 10.3389/fimmu.2020.00163] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Mast cells (MCs) are important sensor and effector cells of the immune system that are involved in many physiological and pathological conditions. Increasing evidence suggests that they also play an important role in bone metabolism and bone disorders. MCs are located in the bone marrow and secrete a wide spectrum of mediators, which can be rapidly released upon activation of mature MCs following their differentiation in mucosal or connective tissues. Many of these mediators can exert osteocatabolic effects by promoting osteoclast formation [e.g., histamine, tumor necrosis factor (TNF), interleukin-6 (IL-6)] and/or by inhibiting osteoblast activity (e.g., IL-1, TNF). By contrast, MCs could potentially act in an osteoprotective manner by stimulating osteoblasts (e.g., transforming growth factor-β) or reducing osteoclastogenesis (e.g., IL-12, interferon-γ). Experimental studies investigating MC functions in physiological bone turnover using MC-deficient mouse lines give contradictory results, reporting delayed or increased bone turnover or no influence depending on the mouse model used. By contrast, the involvement of MCs in various pathological conditions affecting bone is evident. MCs may contribute to the pathogenesis of primary and secondary osteoporosis as well as inflammatory disorders, including rheumatoid arthritis and osteoarthritis, because increased numbers of MCs were found in patients suffering from these diseases. The clinical observations could be largely confirmed in experimental studies using MC-deficient mouse models, which also provide mechanistic insights. MCs also regulate bone healing after fracture by influencing the inflammatory response toward the fracture, vascularization, bone formation, and callus remodeling by osteoclasts. This review summarizes the current view and understanding of the role of MCs on bone in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jochen Kroner
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
29
|
Si J, Wang C, Zhang D, Wang B, Hou W, Zhou Y. Osteopontin in Bone Metabolism and Bone Diseases. Med Sci Monit 2020; 26:e919159. [PMID: 31996665 PMCID: PMC7003659 DOI: 10.12659/msm.919159] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
Osteopontin (OPN), a secreted phosphoprotein, is a member of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family of cell matrix proteins and participates in many biological activities. Studies have shown that OPN plays a role in bone metabolism and homeostasis. OPN not only is an important factor in neuron-mediated and endocrine-regulated bone mass, but also is involved in biological activities such as proliferation, migration, and adhesion of several bone-related cells, including bone marrow mesenchymal stem cells, hematopoietic stem cells, osteoclasts, and osteoblasts. OPN has been demonstrated to be closely related to the occurrence and development of many bone-related diseases, such as osteoporosis, rheumatoid arthritis, and osteosarcoma. As expected, the functions of OPN in the bone have become a research hotspot. In this article, we try to decipher the mechanism of OPN-regulated bone metabolism and bone diseases.
Collapse
Affiliation(s)
- Jinyan Si
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Chaowei Wang
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Denghui Zhang
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Bo Wang
- Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Weiwei Hou
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Yi Zhou
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
30
|
Brioschi S, Zhou Y, Colonna M. Brain Parenchymal and Extraparenchymal Macrophages in Development, Homeostasis, and Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:294-305. [PMID: 31907272 PMCID: PMC7034672 DOI: 10.4049/jimmunol.1900821] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Microglia are parenchymal macrophages of the CNS; as professional phagocytes they are important for maintenance of the brain's physiology. These cells are generated through primitive hematopoiesis in the yolk sac and migrate into the brain rudiment after establishment of embryonic circulation. Thereafter, microglia develop in a stepwise fashion, reaching complete maturity after birth. In the CNS, microglia self-renew without input from blood monocytes. Recent RNA-sequencing studies have defined a molecular signature for microglia under homeostasis. However, during disease, microglia undergo remarkable phenotypic changes, which reflect the acquisition of specialized functions tailored to the pathological context. In addition to microglia, the brain-border regions host populations of extraparenchymal macrophages with disparate origins and phenotypes that have recently been delineated. In this review we outline recent findings that provide a deeper understanding of both parenchymal microglia and extraparenchymal brain macrophages in homeostasis and during disease.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
31
|
Hughes EAB, Robinson TE, Bassett DB, Cox SC, Grover LM. Critical and diverse roles of phosphates in human bone formation. J Mater Chem B 2019; 7:7460-7470. [PMID: 31729501 DOI: 10.1039/c9tb02011j] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Humans utilise biomineralisation in the formation of bone and teeth. Human biomineralisation processes are defined by the transformation of an amorphous phosphate-based precursor to highly organised nanocrystals. Interestingly, ionic phosphate species not only provide a fundamental building block of biological mineral, but rather exhibit several diverse roles in mediating mineral formation in the physiological milieu. In this review, we focus on elucidating the complex roles of phosphate ions and molecules within human biomineralisation pathways, primarily referring to the nucleation and crystallisation of bone mineral.
Collapse
Affiliation(s)
- Erik A B Hughes
- School of Chemical Engineering, University of Birmingham, B15 2TT, UK. and NIHR Surgical Rec and Microbiology Research Centre, Queen Elizabeth Hospital, Birmingham, UK
| | - Thomas E Robinson
- School of Chemical Engineering, University of Birmingham, B15 2TT, UK.
| | - David B Bassett
- School of Chemical Engineering, University of Birmingham, B15 2TT, UK. and Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, B15 2TT, UK.
| | - Liam M Grover
- School of Chemical Engineering, University of Birmingham, B15 2TT, UK.
| |
Collapse
|
32
|
Yaghubi E, Daneshpazhooh M, DJalali M, Mohammadi H, Sepandar F, Fakhri Z, Ghaedi E, Keshavarz SA, Balighi K, Mahmoudi H, Zarei M, Javanbakht MH. Effects of l-carnitine supplementation on cardiovascular and bone turnover markers in patients with pemphigus vulgaris under corticosteroids treatment: A randomized, double-blind, controlled trial. Dermatol Ther 2019; 32:e13049. [PMID: 31369185 DOI: 10.1111/dth.13049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/14/2019] [Accepted: 07/22/2019] [Indexed: 01/12/2023]
Abstract
Pemphigus vulgaris (PV) is a severe, bullous, autoimmune disease of the skin and mucous membranes. Corticosteroids are usually the main core treatment for controlling PV, which could lead to several side effects such as insulin resistance, osteoporosis, and cardiovascular disorders. The aim of this study is to evaluate the protective effects of l-carnitine (LC) supplementation in PV patients under corticosteroid treatment. In this randomized, double-blind, placebo-controlled clinical trial, 48 patients with PV were divided randomly into two groups to receive 2 g LC (n = 24) or a placebo (n = 24) for 8 weeks, respectively. Serum levels of osteopontin (OPN), bone morphogenic protein 4 (BMP4), cystatin C, systolic and diastolic blood pressure, 25 hydroxyvitamin D3, and LC were evaluated at the beginning and at the end of the study. LC supplementation demonstrated a significant increase in serum carnitine (p < .001). In addition, at the end of the trial, LC supplementation significantly decreased serum BMP4 (p = .003), OPN (p = .03), and cystatin C (p = .001) levels. There was no significant effect on blood pressure in comparison with the placebo. During study, no harmful side effects were reported by patients. These findings indicate that LC supplementation significantly leads to favorable changes in OPN, BMP4, and cystatin C in PV patients under corticosteroid therapy. However, further investigations are required to confirm these results.
Collapse
Affiliation(s)
- Elham Yaghubi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Daneshpazhooh
- Autoimmune Bullous Diseases Research Center, Department of Dermatology, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahmoud DJalali
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farnaz Sepandar
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Fakhri
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ehsan Ghaedi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Ali Keshavarz
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Kamran Balighi
- Autoimmune Bullous Diseases Research Center, Department of Dermatology, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamidreza Mahmoudi
- Autoimmune Bullous Diseases Research Center, Department of Dermatology, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahnaz Zarei
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Hassan Javanbakht
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
33
|
Fitter S, Zannettino ACW. Osteopontin in the pathophysiology of obesity: Is Opn a fat cell foe? Obes Res Clin Pract 2019; 12:249-250. [PMID: 29914635 DOI: 10.1016/j.orcp.2018.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Stephen Fitter
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide and the Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide and the Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
34
|
Bouleftour W, Juignet L, Verdière L, Machuca-Gayet I, Thomas M, Laroche N, Vanden-Bossche A, Farlay D, Thomas C, Gineyts E, Concordet JP, Renaud JB, Aubert D, Teixeira M, Peyruchaud O, Vico L, Lafage-Proust MH, Follet H, Malaval L. Deletion of OPN in BSP knockout mice does not correct bone hypomineralization but results in high bone turnover. Bone 2019; 120:411-422. [PMID: 30529011 DOI: 10.1016/j.bone.2018.12.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/29/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022]
Abstract
The two SIBLING (Small Integrin Binding Ligand N-linked Glycoproteins), bone sialoprotein (BSP) and osteopontin (OPN) are expressed in osteoblasts and osteoclasts. In mature BSP knockout (KO, -/-) mice, both bone formation and resorption as well as mineralization are impaired. OPN-/- mice display impaired resorption, and OPN is described as an inhibitor of mineralization. However, OPN is overexpressed in BSP-/- mice, complicating the understanding of their phenotype. We have generated and characterized mice with a double KO (DKO) of OPN and BSP, to try and unravel their respective contributions. Despite the absence of OPN, DKO bones are still hypomineralized. The SIBLING, matrix extracellular phosphoglycoprotein with ASARM motif (MEPE) is highly overexpressed in both BSP-/- and DKO and may impair mineralization through liberation of its ASARM (Acidic Serine-Aspartate Rich MEPE associated) peptides. DKO mice also display evidence of active formation of trabecular, secondary bone as well as primary bone in the marrow-ablation repair model. A higher number of osteoclasts form in DKO marrow cultures, with higher resorption activity, and DKO long bones display a localized and conspicuous cortical macroporosity. High bone formation and resorption parameters, and high cortical porosity in DKO mice suggest an active bone modeling/remodeling, in the absence of two key regulators of bone cell performance. This first double KO of SIBLING proteins thus results in a singular, non-trivial phenotype leading to reconsider the interpretation of each single KO, concerning in particular matrix mineralization and the regulation of bone cell activity.
Collapse
Affiliation(s)
- W Bouleftour
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - L Juignet
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - L Verdière
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | | | - M Thomas
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - N Laroche
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - A Vanden-Bossche
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - D Farlay
- Inserm U1033-Lyos, Université de Lyon, F69372 Lyon, France
| | - C Thomas
- Inserm U1033-Lyos, Université de Lyon, F69372 Lyon, France
| | - E Gineyts
- Inserm U1033-Lyos, Université de Lyon, F69372 Lyon, France
| | - J P Concordet
- Inserm U1154/Cnrs UMR7196/Muséum National d'Histoire Naturelle, F75231 Paris, France
| | - J B Renaud
- Inserm U1154/Cnrs UMR7196/Muséum National d'Histoire Naturelle, F75231 Paris, France
| | - D Aubert
- AniRa PBES, Gerland, F69007 Lyon Sud, France
| | - M Teixeira
- AniRa PBES, Gerland, F69007 Lyon Sud, France
| | - O Peyruchaud
- Inserm U1033-Lyos, Université de Lyon, F69372 Lyon, France
| | - L Vico
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - M H Lafage-Proust
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France
| | - H Follet
- Inserm U1033-Lyos, Université de Lyon, F69372 Lyon, France
| | - L Malaval
- Inserm U1059-Sainbiose, Université de Lyon, F 42270 Saint Priest en Jarez, France.
| |
Collapse
|
35
|
Scholtysek C, Ipseiz N, Böhm C, Krishnacoumar B, Stenzel M, Czerwinski T, Palumbo-Zerr K, Rothe T, Weidner D, Klej A, Stoll C, Distler J, Tuckermann J, Herrmann M, Fabry B, Goldmann WH, Schett G, Krönke G. NR4A1 Regulates Motility of Osteoclast Precursors and Serves as Target for the Modulation of Systemic Bone Turnover. J Bone Miner Res 2018; 33:2035-2047. [PMID: 29949664 DOI: 10.1002/jbmr.3533] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/29/2018] [Accepted: 06/21/2018] [Indexed: 11/10/2022]
Abstract
NR4A1 (Nur77 or NGFI-B), an orphan member of the nuclear receptor superfamily, has been identified as a key regulator of the differentiation and function of myeloid, lymphoid, and mesenchymal cells. The detailed role of NR4A1 in bone biology is incompletely understood. Here, we report a role for NR4A1 as novel factor controlling the migration and recruitment of osteoclast precursors during bone remodeling. Myeloid-specific but not osteoblast-specific deletion of NR4A1 resulted in osteopenia due to an increase in the number of bone-lining osteoclasts. Although NR4A1-deficient osteoclast precursors displayed a regular differentiation into mature osteoclasts, they showed a hyper-motile phenotype that was largely dependent on increased osteopontin expression, suggesting that expression of NR4A1 negatively controlled osteopontin-mediated recruitment of osteoclast precursors to the trabecular bone. Pharmacological activation of NR4A1, in turn, inhibited osteopontin expression and osteopontin-dependent migration of osteoclast precursors resulted in reduced abundance of bone-resorbing osteoclasts in vivo as well as in an ameliorated bone loss after ovariectomy in mice. This study identifies NR4A1 as a crucial player in the regulation of osteoclast biology and bone remodeling and highlights this nuclear receptor as a promising target for therapeutic intervention during the treatment of osteoporosis. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Carina Scholtysek
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Natacha Ipseiz
- School of Medicine, University of Cardiff, Cardiff, Wales
| | - Christina Böhm
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Brenda Krishnacoumar
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Martin Stenzel
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tina Czerwinski
- Department of Biophysics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Katrin Palumbo-Zerr
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tobias Rothe
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Daniela Weidner
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexandra Klej
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Cornelia Stoll
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jörg Distler
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ben Fabry
- Department of Biophysics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang H Goldmann
- Department of Biophysics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
36
|
Icer MA, Gezmen-Karadag M. The multiple functions and mechanisms of osteopontin. Clin Biochem 2018; 59:17-24. [PMID: 30003880 DOI: 10.1016/j.clinbiochem.2018.07.003] [Citation(s) in RCA: 333] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/03/2018] [Accepted: 07/08/2018] [Indexed: 12/12/2022]
Abstract
Osteopontin (OPN) is a highly phosphorylated glycophosphoprotein having acidic characteristics and rich in aspartic acid. OPN, a multifunctional protein, has important functions on cardiovascular diseases, cancer, diabetes and kidney stone diseases and in the process of inflammation, biomineralization, cell viability and wound healing. Osteopontin acts on organisms by playing a key role in secretion levels of interleukin-10 (IL-10), interleukin-12 (IL-12), interleukin-3 (IL-3), interferon-γ (IFN-γ), integrin αvB3, nuclear factor kappa B (NF-kB), macrophage and T cells, regulating the osteoclast function and affecting CD44 receptors. The aim of the present review is to address majority of different functions of OPN protein which are known, suspected or suggested through the data obtained about this protein yet.
Collapse
Affiliation(s)
- Mehmet Arif Icer
- Gazi University, Faculty of Health Sciences, Nutrition and Dietetics Department, 06500 Beşevler, Ankara, Turkey.
| | - Makbule Gezmen-Karadag
- Gazi University, Faculty of Health Sciences, Nutrition and Dietetics Department, 06500 Beşevler, Ankara, Turkey.
| |
Collapse
|
37
|
Mortada I, Mortada R. Dental pulp stem cells and osteogenesis: an update. Cytotechnology 2018; 70:1479-1486. [PMID: 29938380 DOI: 10.1007/s10616-018-0225-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Dental pulp stem cells constitute an attractive source of multipotent mesenchymal stem cells owing to their high proliferation rate and multilineage differentiation potential. Osteogenesis is initiated by osteoblasts, which originate from mesenchymal stem cells. These cells express specific surface antigens that disappear gradually during osteodifferentiation. In parallel, the appearance of characteristic markers, including alkaline phosphatase, collagen type I, osteocalcin and osteopontin characterize the osteoblastic phenotype of dental pulp stem cells. This review will shed the light on the osteogenic differentiation potential of dental pulp stem cells and explore the culture medium components, and markers associated with osteodifferentiation of these cells.
Collapse
|
38
|
Rowe DW, Adams DJ, Hong SH, Zhang C, Shin DG, Renata Rydzik C, Chen L, Wu Z, Garland G, Godfrey DA, Sundberg JP, Ackert-Bicknell C. Screening Gene Knockout Mice for Variation in Bone Mass: Analysis by μCT and Histomorphometry. Curr Osteoporos Rep 2018; 16:77-94. [PMID: 29508144 DOI: 10.1007/s11914-018-0421-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The international mouse phenotyping consortium (IMPC) is producing defined gene knockout mouse lines. Here, a phenotyping program is presented that is based on micro-computed tomography (μCT) assessment of distal femur and vertebra. Lines with significant variation undergo a computer-based bone histomorphometric analysis. RECENT FINDINGS Of the 220 lines examined to date, approximately 15% have a significant variation (high or low) by μCT, most of which are not identified by the IMPC screen. Significant dimorphism between the sexes and bone compartments adds to the complexity of the skeletal findings. The μCT information that is posted at www.bonebase.org can group KOMP lines with similar morphological features. The histological data is presented in a graphic form that associates the cellular features with a specific anatomic group. The web portal presents a bone-centric view appropriate for the skeletal biologist/clinician to organize and understand the large number of genes that can influence skeletal health. Cataloging the relative severity of each variant is the first step towards compiling the dataset necessary to appreciate the full polygenic basis of degenerative bone disease.
Collapse
Affiliation(s)
- David W Rowe
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT, 06030, USA.
| | - Douglas J Adams
- Department of Orthopaedic Surgery, School of Medicine, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Seung-Hyun Hong
- Computer Science and Engineering, School of Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Caibin Zhang
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Dong-Guk Shin
- Computer Science and Engineering, School of Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - C Renata Rydzik
- Department of Orthopaedic Surgery, School of Medicine, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Li Chen
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Zhihua Wu
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT, 06030, USA
| | | | - Dana A Godfrey
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester School of Medicine, Rochester, NY, 14642, USA
| | | | - Cheryl Ackert-Bicknell
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester School of Medicine, Rochester, NY, 14642, USA
| |
Collapse
|
39
|
Kusumi K, Barr-Beare E, Saxena V, Safedi F, Schwaderer A. Renal Calcium Oxalate Deposits Induce a Pro-Atherosclerotic and Pro-Osteoporotic Response in Mice. J Cell Biochem 2017; 118:2744-2751. [PMID: 28176358 DOI: 10.1002/jcb.25924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 11/08/2022]
Abstract
Urinary stone disease (USD) is increasing in adult and pediatric populations. Adult and pediatric studies have demonstrated decreased bone mineral density and increased fracture rates. USD has also been independently linked to increased rates of myocardial infarction and cerebral vascular accidents. Although USD is a multisystem disorder involving the kidneys, bone, and vasculature, the molecular mechanisms linking these three organs remain unknown. Calcium oxalate nephropathy was induced in C57BL/6J mice with intra-peritoneal (ip) injection of sodium glyoxolate. Half of each kidney underwent Pizzalato staining and half was snap frozen for RNA extraction. RT2 Profiler Mouse Atherosclerosis, Osteoporosis, and Calcium Signaling PCR Arrays (Qiagen) were performed. Only results that passed quality checks in PCR array reproducibility and genomic DNA contamination were included. Genes had to show at least fourfold differential expression and P < 0.01 to be considered significant. Atherosclerosis array showed upregulation of 19 genes by fourfold, 10 of which were ≥10-fold. All 19 had P ≤ 0.002. The Osteoporosis array showed fourfold upregulation of 10 genes, five showed >10-fold increase. All 10 have P ≤ 0.003. The calcium signaling array showed significant fourfold upregulation of 10 genes, four of which were ≥10-fold. All 10 have P ≤ 0.03. We have demonstrated that calcium oxalate nephropathy can induce upregulation of atherosclerotic, metabolic bone, and calcium homeostasis genes in a murine model. This may be and initial step in identifying the molecular mechanisms linking stone, bone, and cardiovascular disease. J. Cell. Biochem. 118: 2744-2751, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kirsten Kusumi
- Division of Nephrology, Akron Children's Hospital, Akron, Ohio
| | - Evan Barr-Beare
- The Center for Clinical and Translational Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Vijay Saxena
- The Center for Clinical and Translational Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Fayez Safedi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio
| | - Andrew Schwaderer
- The Center for Clinical and Translational Medicine, Nationwide Children's Hospital, Columbus, Ohio.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital/The Ohio State University, Columbus, Ohio
| |
Collapse
|
40
|
Abstract
Nuclear receptors are a family of transcription factors that can be activated by lipophilic ligands. They are fundamental regulators of development, reproduction, and energy metabolism. In bone, nuclear receptors enable bone cells, including osteoblasts, osteoclasts, and osteocytes, to sense their dynamic microenvironment and maintain normal bone development and remodeling. Our views of the molecular mechanisms in this process have advanced greatly in the past decade. Drugs targeting nuclear receptors are widely used in the clinic for treating patients with bone disorders such as osteoporosis by modulating bone formation and resorption rates. Deficiency in the natural ligands of certain nuclear receptors can cause bone loss; for example, estrogen loss in postmenopausal women leads to osteoporosis and increases bone fracture risk. In contrast, excessive ligands of other nuclear receptors, such as glucocorticoids, can also be detrimental to bone health. Nonetheless, the ligand-induced osteoprotective effects of many other nuclear receptors, e.g., vitamin D receptor, are still in debate and require further characterizations. This review summarizes previous studies on the roles of nuclear receptors in bone homeostasis and incorporates the most recent findings. The advancement of our understanding in this field will help researchers improve the applications of agonists, antagonists, and selective modulators of nuclear receptors for therapeutic purposes; in particular, determining optimal pharmacological drug doses, preventing side effects, and designing new drugs that are more potent and specific.
Collapse
|
41
|
Icariin influences adipogenic differentiation of stem cells affected by osteoblast-osteoclast co-culture and clinical research adipogenic. Biomed Pharmacother 2017; 88:436-442. [PMID: 28122309 DOI: 10.1016/j.biopha.2017.01.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To build mouse osteoblast MC3T3-E1 and mouse osteoclast RAW264.7 co-culture system and to study the effect of icariin on the activity of osteoblasts and osteoclasts in the co-culture system. METHODS In vitro acquisition and cultivation of mouse osteoblasts MC3T3-E1 and mouse RAW264.7 cells were conducted. Osteoblast and osteoclast activities of cells were detected by CCK-8 staining experiment, alizarin red staining and tartaric-resistant acid phosphatase (TRAP) staining. We used different concentrations of icariin to interfere in osteoblast-osteoclast co-culture system. The effects of icariin on various genes were detected by PCR and Western blot methods The correction between the expression of PPARγ and BMD was analyzed in patients with osteoporosis. RESULTS Mouse osteoblast-osteoclast co-culture system was built, and the osteogenic differentiation effect was enhanced. Icariin can improve the MC3T3-E1 osteogenic differentiation activity, enhance the expression of OPG and RANKL gene protein, reduce the NF-κb gene and protein expression, increase of ALP, TGF-b1 and RANKL gene expression level and reduce RANK gene expression. Icariin can act on MC3T3-E1 cells-RAW264.7 cells co-culture system, and promote the osteogenic activity of MC3T3-E1 cells, inhibit the osteoclast activity of RAW264.7 cells and reduce the level of BMSCs adipogenic differentiation. The expression level of PPAR-γ gene was negatively correlated with the level of BMD. CONCLUSIONS Mouse MC3T3-E1 cells and mouse RAW264.7 cells could be co-cultured in vitro, and icariin could improve the osteogenic activity of MC3T3 cells-RAW264.7 cells and decrease the osteoclast activity. Icariin could inhibit adipogenic differentiation of BMSCs in the osteoblast-osteoclast co-culture, promoting osteogenic differentiation and inhibiting osteoclast differentiation.
Collapse
|
42
|
Karakus E, Halici Z, Albayrak A, Bayir Y, Demirci E, Aydin A, Ozturk-Karagoz B, Cadirci E, Ayan AK, Sahin A, Unal D. Effects of Administration of Amlodipine and Lacidipine on Inflammation-Induced Bone Loss in the Ovariectomized Rat. Inflammation 2016; 39:336-346. [PMID: 26412256 DOI: 10.1007/s10753-015-0254-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This study was performed to evaluate the possible protective effect of two calcium channel blocker's "lacidipine (LAC) and amlodipine (AML)" on bone metabolism in an experimental ovariectomized and inflammation-induced osteoporosis rat model (OVXinf). For the purpose of this study, the rats were divided into eight groups, each containing eight rats: sham-operated control (group 1, SH), sham + inflammation (group 2, SHinf), ovariectomy (group 3, OVX), ovariectomy + inflammation (group 4, OVXinf), ovariectomy + LAC 4 mg/kg (group 5, OVX + LAC), ovariectomy + inflammation + LAC 4 mg/kg (group 6, OVXinf + LAC), ovariectomy + AML 5 mg/kg (group 7, OVX + AML), ovariectomy + inflammation + AML 5 mg/kg (group 8, OVXinf + AML). The levels of osteocalcin and osteopontin decreased in OVXinf + LAC and OVXinf + AML groups. The serum levels of TNF-α, IL-1β, and IL-6 were increased significantly in the OVXinf rats compared with the SH group. Gene expression levels of the osteogenic factor runt-related transcription factor 2 (Runx2) and type I collagen 1A1 (Col1A1) significantly decreased in the OVXinf group, when compared with the control group. AML or LAC administrations increased the levels of Runx2 and Col1A1. These results suggest that amlodipine and lacidipine may be a novel therapeutic target for radical osteoporosis treatment in hypertensive patients.
Collapse
Affiliation(s)
- Emre Karakus
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ataturk University, 25240, Erzurum, Turkey.
| | - Zekai Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Abdulmecit Albayrak
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Yasin Bayir
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Elif Demirci
- Department of Pathology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ali Aydin
- Department of Orthopedics and Traumatology, Ataturk University Faculty of Medicine, Erzurum, Turkey
| | - Berna Ozturk-Karagoz
- Department of Pharmacology, Faculty of Pharmacy, Ibrahim Cecen University, Agrı, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Arif Kursat Ayan
- Department of Nuclear Medicine, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ali Sahin
- Department of Nuclear Medicine, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Deniz Unal
- Department of Histology and Embryology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
43
|
Feng JQ, Huang H, Lu Y, Ye L, Xie Y, Tsutsui TW, Kunieda T, Castranio T, Scott G, Bonewald LB, Mishina Y. The Dentin Matrix Protein 1 (Dmp1) is Specifically Expressed in Mineralized, but not Soft, Tissues during Development. J Dent Res 2016; 82:776-80. [PMID: 14514755 DOI: 10.1177/154405910308201003] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Dentin Matrix Protein 1 ( Dmp1) was originally identified from dentin. However, its expression and function in vivo are not clear. To clarify these two issues, we have generated mice carrying a truncated Dmp1 gene by using gene targeting to replace exon 6 with a lacZ gene. Northern blot analysis shows the expected 5.8-kb Dmp1-lacZ fusion transcript and loss of the wild-type 2.8-kb Dmp1 transcript, confirmed by a lack of immunostaining for the protein. Using heterozygous animals, we demonstrate that Dmp1 is specific for mineralized tissues. Not previously shown, Dmp1 is also expressed in pulp cells. Dmp1-deficient embryos and newborns display no apparent gross abnormal phenotype, although there are a modest expansion of the hypertrophic chondrocyte zone and a modest increase in the long bone diameter. This suggests that DMP1 is not essential for early mouse skeletal or dental development.
Collapse
Affiliation(s)
- J Q Feng
- Department of Oral Biology, School of Dentistry, University of Missouri-Kansas City, 650 E. 25th Street, Kansas City, MO 64108, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Carnesecchi J, Vanacker JM. Estrogen-Related Receptors and the control of bone cell fate. Mol Cell Endocrinol 2016; 432:37-43. [PMID: 26206717 DOI: 10.1016/j.mce.2015.07.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/23/2015] [Accepted: 07/17/2015] [Indexed: 11/17/2022]
Abstract
Bone loss is naturally occurring in aging males and females and exacerbated in the latter after menopause, altogether leading to cumulative skeleton fragility and increased fracture risk. Two types of therapeutic strategies can be envisioned to counteract age- or menopause-associated bone loss, aiming at either reducing bone resorption exerted by osteoclasts or, alternatively, promoting bone formation by osteoblasts. We here summarize data suggesting that inhibition of the Estrogen-Related Receptors α and/or γ could promote bone formation and compensate for bone loss induced by ageing or estrogen-deficiency.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
45
|
An J, Yang H, Zhang Q, Liu C, Zhao J, Zhang L, Chen B. Natural products for treatment of osteoporosis: The effects and mechanisms on promoting osteoblast-mediated bone formation. Life Sci 2016; 147:46-58. [DOI: 10.1016/j.lfs.2016.01.024] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 01/04/2016] [Accepted: 01/13/2016] [Indexed: 01/03/2023]
|
46
|
Trotter TN, Yang Y. Matricellular proteins as regulators of cancer metastasis to bone. Matrix Biol 2016; 52-54:301-314. [PMID: 26807761 DOI: 10.1016/j.matbio.2016.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/19/2016] [Accepted: 01/19/2016] [Indexed: 01/08/2023]
Abstract
Metastasis is the major cause of death in cancer patients, and a frequent site of metastasis for many cancers is the bone marrow. Therefore, understanding the mechanisms underlying the metastatic process is necessary for future prevention and treatment. The tumor microenvironment is now known to play a role in the metastatic cascade, both at the primary tumor and in metastatic sites, and includes both cellular and non-cellular components. The extracellular matrix (ECM) provides structural support and signaling cues to cells. One particular group of molecules associated with the ECM, known as matricellular proteins, modulate multiple aspects of tumor biology, including growth, migration, invasion, angiogenesis and metastasis. These proteins are also important for normal function in the bone by regulating bone formation and bone resorption. Recent studies have described a link between some of these proteins and metastasis of various tumors to the bone. The aim of this review is to summarize what is currently known about matricellular protein influence on bone metastasis. Particular attention to the contribution of both tumor cells and non-malignant cells in the bone has been given.
Collapse
Affiliation(s)
- Timothy N Trotter
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yang Yang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States; Comprehensive Cancer Center and the Center for Metabolic Bone Disease, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
47
|
Bouleftour W, Juignet L, Bouet G, Granito RN, Vanden-Bossche A, Laroche N, Aubin JE, Lafage-Proust MH, Vico L, Malaval L. The role of the SIBLING, Bone Sialoprotein in skeletal biology - Contribution of mouse experimental genetics. Matrix Biol 2016; 52-54:60-77. [PMID: 26763578 DOI: 10.1016/j.matbio.2015.12.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/23/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022]
Abstract
Bone Sialoprotein (BSP) is a member of the "Small Integrin-Binding Ligand N-linked Glycoproteins" (SIBLING) extracellular matrix protein family of mineralized tissues. BSP has been less studied than other SIBLING proteins such as Osteopontin (OPN), which is coexpressed with it in several skeletal cell types. Here we review the contribution of genetically engineered mice (BSP gene knockout and overexpression) to the understanding of the role of BSP in the bone organ. The studies made so far highlight the role of BSP in skeletal mineralization, as well as its importance for proper osteoblast and osteoclast differentiation and activity, most prominently in primary/repair bone. The absence of BSP also affects the local environment of the bone tissue, in particular hematopoiesis and vascularization. Interestingly, lack of BSP induces an overexpression of OPN, and the cognate protein could be responsible for some aspects of the BSP gene knockout skeletal phenotype, while replacing BSP for some of its functions. Such interplay between the partly overlapping functions of SIBLING proteins, as well as the network of cross-regulations in which they are involved should now be the focus of further work.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Laura Juignet
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Guenaelle Bouet
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, UK
| | | | - Arnaud Vanden-Bossche
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Norbert Laroche
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marie-Hélène Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Laurence Vico
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Luc Malaval
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France.
| |
Collapse
|
48
|
Abstract
Tilapia type I atelocollagen (TAC) is a strong candidate for clinical application as its biological scaffold due to a high degeneration temperature and biologically safe properties. The aim of this study was to confirm the biological effects of TACin vitroon osteoblastic cells, simulating its clinical application. The proliferation and differentiation of typical preosteoblasts, MC3T3-E1 cells, were investigated using a microarray analysis, staining assay for mineralization, and real-time PCR analysis of the expression of mineralization-related genes. The mRNA expression of 10 genes involved in proliferation and differentiation increased after 3-day culture on an TAC gel, with an average balanced score ratio exceeding 1.5 compared to the control. After two weeks of culture, all three experimental groups showed stronger alkaline phosphatase staining than after one week. The genes expression of alkaline phosphatase, osteocalcin, and bone sialoprotein increased under the experimental conditions. The gene expression of osteopontin did not increase, and no statistical differences were noted among the three experimental groups. The present and previous findings suggest that TAC is not only a suitable alternative to collagen products originating from mammals but also a novel biomaterial with cell differentiation ability for regenerative medicine.
Collapse
|
49
|
Wei QS, Wang HB, Wang JL, Fang B, Zhou GQ, Tan X, He W, Deng WM. Combination treatment with whole body vibration and a kidney-tonifying herbal Fufang prevent osteoporosis in ovariectomized rats. Orthop Surg 2015; 7:57-65. [PMID: 25708037 DOI: 10.1111/os.12161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/30/2014] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE To assess the ability of whole body vibration (WBV) with the kidney-tonifying herbal Fufang (Bushen Zhuanggu Granules, BZG) to prevent osteoporosis in ovariectomized rats. METHODS Fifty 6-month-old female Sprague Dawley rats were divided into five groups: sham-operated (SHAM), ovariectomized (OVX), OVX with WBV (OVX + WBV), OVX with BZG (OVX + BZG), OVX with both WBV and BZG (OVX + WBV + BZG). The SHAM group received normal saline. After 12 weeks of treatment, the rats were killed, their serum concentrations of osteopontin (OPN), receptor activator of nuclear factor kappa-B ligand RANKL and bone turnover markers assayed and bone mineral density (BMD), histomorphometry and bone strength evaluated. RESULTS Concentrations of OPN were significantly lower in the SHAM, OVX + WBV and OVX + WBV + BZG groups at 12 weeks, whereas concentrations of RANKL had decreased significantly in the SHAM, OVX + WBV, OVX + BZG and OVX + WBV + BZG groups. In the OVX + WBV, OVX + BZG and OVX + WBV + BZG groups the amount of bone turnover had been significantly antagonized. Compared with OVX group, BMD, % trabecular area (Tb.Ar), number of trabeculae (Tb.N) and assessed biomechanical variables were higher in OVX+WBV group, whereas and BMD, %Tb.Ar, Tb.N, maximal load and yield load were higher in the OVX + BZG group. All tested indices were significantly lower in the OVX + WBV and OVX + BZG groups than in the OVX + WBV + BZG group. CONCLUSION Either WBV or BZG alone prevents OVX-induced bone loss. However, BZG enhances the effect of WBV by further enhancing BMD, bone architecture and strength.
Collapse
Affiliation(s)
- Qiu-shi Wei
- Department of Rehabilitation, General Hospital of Guangzhou Military Command of People's Liberation Army, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
CD44 deficiency inhibits unloading-induced cortical bone loss through downregulation of osteoclast activity. Sci Rep 2015; 5:16124. [PMID: 26530337 PMCID: PMC4632082 DOI: 10.1038/srep16124] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/09/2015] [Indexed: 12/13/2022] Open
Abstract
The CD44 is cellular surface adhesion molecule that is involved in physiological processes such as hematopoiesis, lymphocyte homing and limb development. It plays an important role in a variety of cellular functions including adhesion, migration, invasion and survival. In bone tissue, CD44 is widely expressed in osteoblasts, osteoclasts and osteocytes. However, the mechanisms underlying its role in bone metabolism remain unclear. We found that CD44 expression was upregulated during osteoclastogenesis. CD44 deficiency in vitro significantly inhibited osteoclast activity and function by regulating the NF-κB/NFATc1-mediated pathway. In vivo, CD44 mRNA levels were significantly upregulated in osteoclasts isolated from the hindlimb of tail-suspended mice. CD44 deficiency can reduce osteoclast activity and counteract cortical bone loss in the hindlimb of unloaded mice. These results suggest that therapeutic inhibition of CD44 may protect from unloading induced bone loss by inhibiting osteoclast activity.
Collapse
|