1
|
Li F, Cui S. Knockdown of C3aR alleviates age-related bone loss via activation of YAP1/β-catenin signaling. J Biol Chem 2025; 301:108500. [PMID: 40216253 DOI: 10.1016/j.jbc.2025.108500] [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: 10/12/2024] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 05/12/2025] Open
Abstract
The complement system plays an important role in bone growth during physiological development and skeletal homeostasis. However, the specific impact of the complement C3a receptor (C3aR) on age-related bone loss remains unclear. In this study, we found that C3aR expression increased with age and was the same as that of the senescent molecules p53, p21, and p16 in control mice. Knockdown of C3aR reduced the expression of senescence markers and significantly ameliorated bone senescence. Notably, C3aR knockdown in mice effectively reversed age-induced bone loss, which was characterized by an increase in the number of osteoblasts and a decrease in the number of osteoclasts. In an in vitro model of D-gal-induced senescence, increased expression of C3aR correlated with increased expression of senescence markers such as p53, p21, and p16. Treatment with a C3aR antagonist (JR14a) successfully attenuated the expression of these markers of cellular senescence and reduced the proportion of late apoptotic cells. Mechanistically, JR14a treatment mitigated D-gal-mediated inhibition of osteoblastic differentiation in preosteoblasts through activation of the YAP1/β-catenin signaling pathway. In the D-gal-induced aging mouse model, treatment with JR14a ameliorates bone microarchitecture and bone loss. In summary, these studies revealed a role for C3aR in regulating bone homeostasis, suggesting that targeting C3aR may be a promising therapeutic strategy for the treatment of age-related osteoporosis.
Collapse
Affiliation(s)
- Fangyu Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shun Cui
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Aparicio-Bautista DI, Becerra-Cervera A, Rivera-Paredez B, Aguilar-Ordoñez I, Ríos-Castro E, Reyes-Grajeda JP, Salmerón J, Hidalgo-Bravo A, Velázquez-Cruz R. Label-free quantitative proteomics in serum reveals candidate biomarkers associated with low bone mineral density in Mexican postmenopausal women. GeroScience 2024; 46:2177-2195. [PMID: 37874452 PMCID: PMC10828159 DOI: 10.1007/s11357-023-00977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/07/2023] [Indexed: 10/25/2023] Open
Abstract
Postmenopausal osteoporosis is a public health problem leading to an increased risk of fractures, negatively impacting women's health. The absence of sensitive and specific biomarkers for early detection of osteoporosis represents a substantial challenge for improving patient management. Herein, we aimed to identify potential candidate proteins associated with low bone mineral density (BMD) in postmenopausal women from the Mexican population. Serum samples from postmenopausal women (40 with normal BMD, 40 with osteopenia (OS), and 20 with osteoporosis (OP)) were analyzed by label-free LC-MS/MS quantitative proteomics. Proteome profiling revealed significant differences between the OS and OP groups compared to individuals with normal BMD. A quantitative comparison of proteins between groups indicated 454 differentially expressed proteins (DEPs). Compared to normal BMD, 14 and 214 DEPs were found in OS and OP groups, respectively, while 226 DEPs were identified between OS and OP groups. The protein-protein interaction and enrichment analysis of DEPs were closely linked to the bone mineral content, skeletal morphology, and immune response activation. Based on their role in bone metabolism, a panel of 12 candidate biomarkers was selected, of which 1 DEP (RYR1) was found upregulated in the OS and OP groups, 8 DEPs (APOA1, SHBG, FETB, MASP1, PTK2B, KNG1, GSN, and B2M) were upregulated in OP and 3 DEPs (APOA2, RYR3, and HBD) were downregulated in OS or OP. The proteomic analysis described here may help discover new and potentially non-invasive biomarkers for the early diagnosis of osteoporosis in postmenopausal women.
Collapse
Affiliation(s)
- Diana I Aparicio-Bautista
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Ciudad de Mexico, Mexico
| | - Adriana Becerra-Cervera
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Ciudad de Mexico, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), 03940, Ciudad de Mexico, Mexico
| | - Berenice Rivera-Paredez
- Centro de Investigación en Políticas, Población y Salud, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico
| | - Israel Aguilar-Ordoñez
- Departamento de Supercómputo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Ciudad de Mexico, Mexico
| | - Emmanuel Ríos-Castro
- Unidad de Genómica, Proteómica y Metabolómica (UGPM), LaNSE, Cinvestav-IPN, 07360, Ciudad de Mexico, Mexico
| | - Juan P Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Ciudad de Mexico, Mexico
| | - Jorge Salmerón
- Centro de Investigación en Políticas, Población y Salud, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico
| | - Alberto Hidalgo-Bravo
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación, 14389, Ciudad de México, Mexico.
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Ciudad de Mexico, Mexico.
| |
Collapse
|
3
|
Meng Q, Wang Y, Yuan T, Su Y, Li Z, Sun S. Osteoclast: The novel whistleblower in osteonecrosis of the femoral head. GENE REPORTS 2023; 33:101833. [DOI: 10.1016/j.genrep.2023.101833] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Gibson BG, Cox TE, Marchbank KJ. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev 2023; 313:194-216. [PMID: 36203396 PMCID: PMC10092198 DOI: 10.1111/imr.13141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review aimed to capture the key findings that animal models have provided around the role of the alternative pathway and amplification loop (AP/AL) in disease. Animal models, particularly mouse models, have been incredibly useful to define the role of complement and the alternative pathway in health and disease; for instance, the use of cobra venom factor and depletion of C3 provided the initial insight that complement was essential to generate an appropriate adaptive immune response. The development of knockout mice have further underlined the importance of the AP/AL in disease, with the FH knockout mouse paving the way for the first anti-complement drugs. The impact from the development of FB, properdin, and C3 knockout mice closely follows this in terms of mechanistic understanding in disease. Indeed, our current understanding that complement plays a role in most conditions at one level or another is rooted in many of these in vivo studies. That C3, in particular, has roles beyond the obvious in innate and adaptive immunity, normal physiology, and cellular functions, with or without other recognized AP components, we would argue, only extends the reach of this arm of the complement system. Humanized mouse models also continue to play their part. Here, we argue that the animal models developed over the last few decades have truly helped define the role of the AP/AL in disease.
Collapse
Affiliation(s)
- Beth G. Gibson
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Thomas E. Cox
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| |
Collapse
|
5
|
Gao Y, Min Q, Li X, Liu L, Lv Y, Xu W, Liu X, Wang H. Immune System Acts on Orthodontic Tooth Movement: Cellular and Molecular Mechanisms. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9668610. [PMID: 36330460 PMCID: PMC9626206 DOI: 10.1155/2022/9668610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/05/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022]
Abstract
Orthodontic tooth movement (OTM) is a tissue remodeling process based on orthodontic force loading. Compressed periodontal tissues have a complicated aseptic inflammatory cascade, which are considered the initial factor of alveolar bone remodeling. Since skeletal and immune systems shared a wide variety of molecules, osteoimmunology has been generally accepted as an interdisciplinary field to investigate their interactions. Unsurprisingly, OTM is considered a good mirror of osteoimmunology since it involves immune reaction and bone remolding. In fact, besides bone remodeling, OTM involves cementum resorption, soft tissue remodeling, orthodontic pain, and relapse, all correlated with immune cells and/or immunologically active substance. The aim of this paper is to review the interaction of immune system with orthodontic tooth movement, which helps gain insights into mechanisms of OTM and search novel method to short treatment period and control complications.
Collapse
Affiliation(s)
- Yajun Gao
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Qingqing Min
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Xingjia Li
- Department of Prosthodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Linxiang Liu
- Department of Implantology, Wuxi Stomatology Hospital, Wuxi, China
| | - Yangyang Lv
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | - Wenjie Xu
- Department of Endodontics, Wuxi Stomatology Hospital, Wuxi, China
| | | | - Hua Wang
- Wuhu Stomatology Hospital, Wuhu, China
| |
Collapse
|
6
|
Çiftci Dede E, Korkusuz P, Bilgiç E, Çetinkaya MA, Korkusuz F. Boron Nano-hydroxyapatite Composite Increases the Bone Regeneration of Ovariectomized Rabbit Femurs. Biol Trace Elem Res 2022; 200:183-196. [PMID: 33715074 DOI: 10.1007/s12011-021-02626-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/03/2021] [Indexed: 10/21/2022]
Abstract
Osteoporosis is a systemic metabolic disease defined by a decreased bone mineral density, microarchitectural deterioration, and an increased incidence of fragility fractures that may lead to morbidity and mortality. Boron may stimulate new bone formation and regeneration, when combined with nano-hydroxyapatite. We questioned whether injecting boron-containing nano-hydroxyapatite composites with hyaluronan increased the bone mineral density and new bone formation in osteoporotic rabbit femurs. The regenerative effects of injectable boron-containing nano-hydroxyapatite composites from 6 to 12 weeks, which may prevent osteoporotic femoral fractures, were assessed. Boron-containing (10 μg/ml) nano-hydroxyapatite composites were injected into the intramedullary femoral cavity with hyaluronan. These significantly increased the histomorphometric new bone surface to the total bone surface ratio at 6 and 9 weeks. The micro-tomographic bone volume to the total volume ratio and bone mineral density in osteoporotic rabbit femurs increased when compared to the hyaluronan (p = 0.004, p = 0.004, p = 0.004, p = 0.01, respectively) and the sham-control (p = 0.01, p = 0.004, p = 0.01, p = 0.037, respectively) groups. The boron-containing group had a higher bone mineralization and new bone formation compared to the nano-hydroxyapatite group, although the difference was not statistically significant. These findings reveal that intramedullary injection of boron-containing nano-hydroxyapatite with hyaluronan increases new bone formation and mineralization in ovariectomized rabbit femurs. Boron-containing nano-hydroxyapatite composites are promising tissue engineering biomaterials that may have regenerative potential in preventing primary and/or secondary femoral fractures in osteoporosis patients.
Collapse
Affiliation(s)
- Eda Çiftci Dede
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, 06810, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye
| | - Elif Bilgiç
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye
| | - Mehmet Alper Çetinkaya
- Animal Research Center, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, 06100, Turkey
| | - Feza Korkusuz
- Department of Sport Medicine, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye.
| |
Collapse
|
7
|
Bülow JM, Renz N, Haffner-Luntzer M, Fischer V, Schoppa A, Tuckermann J, Köhl J, Huber-Lang M, Ignatius A. Complement receptor C5aR1 on osteoblasts regulates osteoclastogenesis in experimental postmenopausal osteoporosis. Front Endocrinol (Lausanne) 2022; 13:1016057. [PMID: 36246887 PMCID: PMC9561253 DOI: 10.3389/fendo.2022.1016057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
In recent years, evidence has accumulated that the complement system, an integral part of innate immunity, may be involved in the regulation of bone homeostasis as well as inflammatory bone loss, for example, in rheumatoid arthritis and periodontitis. Complement may also contribute to osteoporosis development, but investigation of the mechanism is limited. Using mice with a conditional deletion of the complement anaphylatoxin receptor C5aR1, we here demonstrated that C5aR1 in osteoblasts (C5aR1 Runx2-Cre mice) or osteoclasts (C5aR1 LysM-Cre mice) did not affect physiological bone turnover or age-related bone loss in either sex, as confirmed by micro-computed tomography, histomorphometry, and biomechanical analyses of the bone and by the measurement of bone turnover markers in the blood serum. When female mice were subjected to ovariectomy (OVX), a common model for postmenopausal osteoporosis, significant bone loss was induced in C5aR1 fl/fl and C5aR1 LysM-Cre mice, as demonstrated by a significantly reduced bone volume fraction, trabecular number and thickness as well as an increased trabecular separation in the trabecular bone compartment. Confirming this, the osteoclast number and the receptor activator of nuclear factor k-B (RANK) ligand (RANKL) serum level were significantly elevated in these mouse lines. By contrast, C5aR1 Runx2-Cre mice were protected from bone loss after OVX and the serum RANKL concentration was not increased after OVX. These data suggested that bone cell-specific C5aR1 may be redundant in bone homeostasis regulation under physiological conditions. However, C5aR1 on osteoblasts was crucial for the induction of bone resorption under osteoporotic conditions by stimulating RANKL release, whereas C5aR1 on osteoclasts did not regulate OVX-induced bone loss. Therefore, our results implicate C5aR1 on osteoblasts as a potential target for treating postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Jasmin Maria Bülow
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Nikolai Renz
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
- *Correspondence: Anita Ignatius,
| |
Collapse
|
8
|
Aonuma F, Higashi S, Tsuka S, Ohsumi T, Masaki C, Hosokawa R, Takeuchi H. Effect of aging on bone metabolism: the involvement of complement C1q. J Prosthodont Res 2021; 65:393-399. [PMID: 33116027 DOI: 10.2186/jpr.jpor_2019_644] [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] [Indexed: 11/06/2022]
Abstract
Purpose Impairment of normal bone remodeling affects the successful osseointegration of dental implants. Recently, it has been reported that complement C1q level increases with age and delays wound healing by modulating Wnt signaling. As Wnt signaling is known to play an essential role in bone remodeling, we hypothesized that aging-dependent increases in C1q affect bone remodeling. In this study, we examined whether C1q affects the differentiation of bone-forming osteoblasts and bone-resorbing osteoclasts, and investigated whether C1q could modify cellular signaling, including the Wnt/β-catenin pathway in these cells.Methods Osteogenic differentiation of MC3T3-E1 cells was assessed using alkaline phosphatase staining. Differentiation of osteoclasts from mouse bone marrow cells was assessed using tartrate-resistant acid phosphatase staining. Activation of canonical Wnt signaling and protein phosphorylation was monitored using Western blotting.Results C1q, at 5-15 µg/mL promoted osteoclast fusion, whereas it did not affect the differentiation of osteoblasts. On the other hand, a higher concentration of C1q (50 µg/mL) suppressed both bone morphogenetic protein-2-induced osteogenic differentiation and osteoclast formation. C1q did not induce an obvious activation of Wnt/ β-catenin signaling in either pre-osteoblasts or pre-osteoclasts, contrary to previous reports using other tissues. Instead, C1q upregulated the receptor activator of nuclear factor-kappa B ligand (RANKL)-induced phosphorylation of Akt.Conclusions C1q could affect cellular signaling and modify the differentiation of osteoblasts and osteoclasts, depending on the concentration. Therefore, an increase in C1q with age could be one of the factors that determine the prognosis of treatment of elderly patients.
Collapse
Affiliation(s)
- Fumiko Aonuma
- Division of Applied Pharmacology, Kyushu Dental University, Kitakyushu, Japan.,Department of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Japan
| | - Sen Higashi
- Division of Applied Pharmacology, Kyushu Dental University, Kitakyushu, Japan
| | - Shintaro Tsuka
- Department of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Japan
| | - Tomoko Ohsumi
- Division of Applied Pharmacology, Kyushu Dental University, Kitakyushu, Japan
| | - Chihiro Masaki
- Department of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Japan
| | - Ryuji Hosokawa
- Department of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Japan
| | - Hiroshi Takeuchi
- Division of Applied Pharmacology, Kyushu Dental University, Kitakyushu, Japan
| |
Collapse
|
9
|
Jiang F, Liu H, Peng F, Liu Z, Ding K, Song J, Li L, Chen J, Shao Q, Yan S, De Veirman K, Vanderkerken K, Fu R. Complement C3a activates osteoclasts by regulating the PI3K/PDK1/SGK3 pathway in patients with multiple myeloma. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0430. [PMID: 33960177 PMCID: PMC8330530 DOI: 10.20892/j.issn.2095-3941.2020.0430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/27/2020] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Myeloma bone disease (MBD) is the most common complication of multiple myeloma (MM). Our previous study showed that the serum levels of C3/C4 in MM patients were significantly positively correlated with the severity of bone disease. However, the mechanism of C3a/C4a in osteoclasts MM patients remains unclear. METHODS The formation and function of osteoclasts were analyzed after adding C3a/C4a in vitro. RNA-seq analysis was used to screen the potential pathways affecting osteoclasts, and the results were verified by Western blot, qRT-PCR, and pathway inhibitors. RESULTS The osteoclast area per view induced by 1 μg/mL (mean ± SD: 50.828 ± 12.984%) and 10 μg/mL (53.663 ± 12.685%) of C3a was significantly increased compared to the control group (0 μg/mL) (34.635 ± 8.916%) (P < 0.001 and P < 0.001, respectively). The relative mRNA expressions of genes, OSCAR/TRAP/RANKL/cathepsin K, induced by 1 μg/mL (median: 5.041, 3.726, 1.638, and 4.752, respectively) and 10 μg/mL (median: 5.140, 3.702, 2.250, and 5.172, respectively) of C3a was significantly increased compared to the control group (median: 3.137, 2.004, 0.573, and 2.257, respectively) (1 μg/mL P = 0.001, P = 0.003, P < 0.001, and P = 0.008, respectively; 10 μg/mL: P < 0.001, P = 0.019, P < 0.001, and P = 0.002, respectively). The absorption areas of the osteoclast resorption pits per view induced by 1 μg/mL (mean ± SD: 51.464 ± 11.983%) and 10 μg/mL (50.219 ± 12.067%) of C3a was also significantly increased (33.845 ± 8.331%) (P < 0.001 and P < 0.001, respectively) compared to the control. There was no difference between the C4a and control groups. RNA-seq analysis showed that C3a promoted the proliferation of osteoclasts using the phosphoinositide 3-kinase (PI3K) signaling pathway. The relative expressions of PIK3CA/phosphoinositide dependent kinase-1 (PDK1)/serum and glucocorticoid inducible protein kinases (SGK3) genes and PI3K/PDK1/p-SGK3 protein in the C3a group were significantly higher than in the control group. The activation role of C3a in osteoclasts of MM patients was reduced by the SGK inhibitor (EMD638683). CONCLUSIONS C3a activated osteoclasts by regulating the PI3K/PDK1/SGK3 pathways in MM patients, which was reduced using a SGK inhibitor. Overall, our results identified potential therapeutic targets and strategies for MBD patients.
Collapse
Affiliation(s)
- Fengjuan Jiang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fengping Peng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jin Chen
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qing Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Siyang Yan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kim De Veirman
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
10
|
Liu Y, Bi Y, Chai L, Song L, Huang J, Wang Q, Li Y, Zhou K. Development of epimedin A complex drugs for treating the osteoporosis. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:17. [PMID: 33506368 PMCID: PMC7840628 DOI: 10.1007/s10856-020-06472-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
Osteoporosis is the most common disease involving bone degeneration. As the age of the population increases, the prevalence of the disease is expected to rise. However, current treatment methods do not provide a desirable solution for the restoration of the function of degenerated bones in patients with osteoporosis. This led to emergence of controlled delivery systems to increase drug bioavailability and efficacy specifically at the bone regeneration. In this study, an epimedin A (EA) complex drug system was prepared by solution blending method. In vitro cell-based experiments showed that the EA complex drug could significantly promote the differentiation and proliferation of osteoblasts and increase the alkaline phosphatase activity, calcium nodule formation, and the expression of osteogenesis-related genes and proteins. In vivo experiments further demonstrated that this novel drugs remarkably enhanced bone regeneration. These results suggest that EA may be used for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Ying Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yanan Bi
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Lijuan Chai
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, 301617, China
| | - Lei Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Juyang Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qin Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Yunzhang Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| | - Kun Zhou
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
11
|
Aaron N, Kraakman MJ, Zhou Q, Liu Q, Costa S, Yang J, Liu L, Yu L, Wang L, He Y, Fan L, Hirakawa H, Ding L, Lo J, Wang W, Zhao B, Guo E, Sun L, Rosen CJ, Qiang L. Adipsin promotes bone marrow adiposity by priming mesenchymal stem cells. eLife 2021; 10:69209. [PMID: 34155972 PMCID: PMC8219379 DOI: 10.7554/elife.69209] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/07/2021] [Indexed: 01/12/2023] Open
Abstract
Background Marrow adipose tissue (MAT) has been shown to be vital for regulating metabolism and maintaining skeletal homeostasis in the bone marrow (BM) niche. As a reflection of BM remodeling, MAT is highly responsive to nutrient fluctuations, hormonal changes, and metabolic disturbances such as obesity and diabetes mellitus. Expansion of MAT has also been strongly associated with bone loss in mice and humans. However, the regulation of BM plasticity remains poorly understood, as does the mechanism that links changes in marrow adiposity with bone remodeling. Methods We studied deletion of Adipsin, and its downstream effector, C3, in C57BL/6 mice as well as the bone-protected PPARγ constitutive deacetylation 2KR mice to assess BM plasticity. The mice were challenged with thiazolidinedione treatment, calorie restriction, or aging to induce bone loss and MAT expansion. Analysis of bone mineral density and marrow adiposity was performed using a μCT scanner and by RNA analysis to assess adipocyte and osteoblast markers. For in vitro studies, primary bone marrow stromal cells were isolated and subjected to osteoblastogenic or adipogenic differentiation or chemical treatment followed by morphological and molecular analyses. Clinical data was obtained from samples of a previous clinical trial of fasting and high-calorie diet in healthy human volunteers. Results We show that Adipsin is the most upregulated adipokine during MAT expansion in mice and humans in a PPARγ acetylation-dependent manner. Genetic ablation of Adipsin in mice specifically inhibited MAT expansion but not peripheral adipose depots, and improved bone mass during calorie restriction, thiazolidinedione treatment, and aging. These effects were mediated through its downstream effector, complement component C3, to prime common progenitor cells toward adipogenesis rather than osteoblastogenesis through inhibiting Wnt/β-catenin signaling. Conclusions Adipsin promotes new adipocyte formation and affects skeletal remodeling in the BM niche. Our study reveals a novel mechanism whereby the BM sustains its own plasticity through paracrine and endocrine actions of a unique adipokine. Funding This work was supported by the National Institutes of Health T32DK007328 (NA), F31DK124926 (NA), R01DK121140 (JCL), R01AR068970 (BZ), R01AR071463 (BZ), R01DK112943 (LQ), R24DK092759 (CJR), and P01HL087123 (LQ).
Collapse
Affiliation(s)
- Nicole Aaron
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pharmacology, Columbia UniversityNew YorkUnited States
| | - Michael J Kraakman
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Qiongming Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States,School of Medicine, Tufts UniversityBostonUnited States,Graduate School of Biomedical Science and Engineering, University of MaineOronoUnited States
| | - Jing Yang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Longhua Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lexiang Yu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Liheng Wang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Ying He
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lihong Fan
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Hiroyuki Hirakawa
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Lei Ding
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - James Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, and Division of Cardiology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science CenterOklahoma CityUnited States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and The David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, Department of Medicine, Weill Cornell Medical College; Graduate Program in Cell & Developmental Biology, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Edward Guo
- Department of Biomedical Engineering, Columbia UniversityNew YorkUnited States
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Cliff J Rosen
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States
| | - Li Qiang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
12
|
Abstract
Objectives The aim of this study was to provide a comprehensive understanding of alterations in messenger RNAs (mRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) in cartilage affected by osteoarthritis (OA). Methods The expression profiles of mRNAs, lncRNAs, and circRNAs in OA cartilage were assessed using whole-transcriptome sequencing. Bioinformatics analyses included prediction and reannotation of novel lncRNAs and circRNAs, their classification, and their placement into subgroups. Gene ontology and pathway analysis were performed to identify differentially expressed genes (DEGs), differentially expressed lncRNAs (DELs), and differentially expressed circRNAs (DECs). We focused on the overlap of DEGs and targets of DELs previously identified in seven high-throughput studies. The top ten DELs were verified by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in articular chondrocytes, both in vitro and in vivo. Results In total, 739 mRNAs, 1152 lncRNAs, and 42 circRNAs were found to be differentially expressed in OA cartilage tissue. Among these, we identified 18 overlapping DEGs and targets of DELs, and the top ten DELs were screened by expression profile analysis as candidate OA-related genes. WISP2, ATF3, and CHI3L1 were significantly increased in both normal versus OA tissues and normal versus interleukin (IL)-1β-induced OA-like cell models, while ADAM12, PRELP, and ASPN were shown to be significantly decreased. Among the identified DELs, we observed higher expression of ENST00000453554 and MSTRG.99593.3, and lower expression of MSTRG.44186.2 and NONHSAT186094.1 in normal versus OA cells and tissues. Conclusion This study revealed expression patterns of coding and noncoding RNAs in OA cartilage, which added sets of genes and noncoding RNAs to the list of candidate diagnostic biomarkers and therapeutic agents for OA patients. Cite this article: H. Li, H. H. Yang, Z. G. Sun, H. B. Tang, J. K. Min. Whole-transcriptome sequencing of knee joint cartilage from osteoarthritis patients. Bone Joint Res 2019;8:290–303. DOI: 10.1302/2046-3758.87.BJR-2018-0297.R1.
Collapse
Affiliation(s)
- H Li
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| | - H H Yang
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| | - Z G Sun
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| | - H B Tang
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| | - J K Min
- Department of Orthopaedics, The First People's Hospital of Huzhou, The First Affiliated Hospital of Huzhou Teachers College, Huzhou, China
| |
Collapse
|
13
|
Connexin 43 hemichannels protect bone loss during estrogen deficiency. Bone Res 2019; 7:11. [PMID: 31016065 PMCID: PMC6476886 DOI: 10.1038/s41413-019-0050-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 02/02/2023] Open
Abstract
Estrogen deficiency in postmenopausal women is a major cause of bone loss, resulting in osteopenia, osteoporosis, and a high risk for bone fracture. Connexin 43 (Cx43) hemichannels (HCs) in osteocytes play an important role in osteocyte viability, bone formation, and remodeling. We showed here that estrogen deficiency reduced Cx43 expression and HC function. To determine if functional HCs protect osteocytes and bone loss during estrogen deficiency, we adopted an ovariectomy model in wild-type (WT) and two transgenic Cx43 mice: R76W (dominant-negative mutant inhibiting only gap junction channels) and Cx43 Δ130–136 (dominant-negative mutant compromising both gap junction channels and HCs). The bone mineral density (BMD), bone structure, and histomorphometric changes of cortical and trabecular bones after ovariectomy were investigated. Our results showed that the Δ130–136 transgenic cohort had greatly decreased vertebral trabecular bone mass compared to WT and R76W mice, associated with a significant increase in the number of apoptotic osteocyte and empty lacunae. Moreover, osteoclast surfaces in trabecular and cortical bones were increased after ovariectomy in the R76W and WT mice, respectively, but not in ∆130–136 mice. These data demonstrate that impairment of Cx43 HCs in osteocytes accelerates vertebral trabecular bone loss and increase in osteocyte apoptosis, and further suggest that Cx43 HCs in osteocytes protect trabecular bone against catabolic effects due to estrogen deficiency. Channels that form between cells and their extracellular environment help protect bone tissue from the damage wrought by low estrogen levels, a major cause of bone loss in post-menopausal women. Jean Jiang from the UT Health San Antonio and colleagues showed that depleting the estrogen hormone in mouse bone cells reduced levels of connexin 43 and impaired the protein’s ability to forms pores known as ‘hemichannels’. The researchers surgically removed the ovaries of various mouse strains to induce estrogen deficiencies. They found that transgenic mice without working hemichannels had reduced bone mass compared to normal mice or mice that could make hemichannels but lacked the ability for those channels to come together to form complete passageways. The findings highlight the importance of connexin 43 hemichannels in protecting bone tissue against osteoporosis.
Collapse
|
14
|
Mödinger Y, Löffler B, Huber-Lang M, Ignatius A. Complement involvement in bone homeostasis and bone disorders. Semin Immunol 2018; 37:53-65. [DOI: 10.1016/j.smim.2018.01.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
|