1
|
Gu H, Pan Y, Xiao H, Zhao L, Tang Y, Ge W. Knockdown of LAP2α inhibits adipogenesis of human adipose-derived stem cells and ameliorates high-fat diet-induced obesity. FASEB J 2024; 38:e23664. [PMID: 38775797 DOI: 10.1096/fj.202302435rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
Adipogenesis, a pivotal cellular process involving the differentiation of mesenchymal stem cells (MSCs) to mature adipocytes, plays a significant role in various physiological functions. Dysregulation of adipogenesis is implicated in conditions such as obesity. However, the complete molecular understanding of adipogenesis remains elusive. This study aimed to uncover the novel role of lamina-associated polypeptide 2 alpha (LAP2α) in human adipose-derived stem cells (hASCs) adipogenesis and its impact on high-fat diet (HFD)-induced obesity and associated metabolic disturbances. LAP2α expression was assessed during the adipogenic differentiation of hASCs using RT-qPCR and western blotting. The functional role of LAP2α in adipogenesis was explored both in vitro and in vivo through loss- and gain-of-function studies. Moreover, mice with HFD-induced obesity received lentivirus injection to assess the effect of LAP2α knockdown on fat accumulation. Molecular mechanisms underlying LAP2α in adipogenic differentiation were investigated using RT-qPCR, Western blotting, immunofluorescence staining, and Oil Red O staining. LAP2α expression was upregulated during hASCs adipogenic differentiation. LAP2α knockdown hindered adipogenesis, while LAP2α overexpression promoted adipogenic differentiation. Notably, LAP2α deficiency resisted HFD-induced obesity, improved glucose intolerance, mitigated insulin resistance, and prevented fatty liver development. Mechanistically, LAP2α knockdown attenuated signal transducer and activator of transcription 3 (STAT3) activation by reducing the protein level of phosphorylated STAT3. A STAT3 activator (Colivelin) counteracted the negative impact of LAP2α deficiency on hASCs adipogenic differentiation. Taken together, our current study established LAP2α as a crucial regulator of hASCs adipogenic differentiation, unveiling a new therapeutic target for obesity prevention.
Collapse
Affiliation(s)
- Hang Gu
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, P.R. China
| | - Yuan Pan
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, P.R. China
| | - Han Xiao
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, P.R. China
| | - Lijun Zhao
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, P.R. China
| | - Yiman Tang
- Fourth Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, P.R. China
| | - Wenshu Ge
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, P.R. China
| |
Collapse
|
2
|
Zhang L, Jiao G, You Y, Li X, Liu J, Sun Z, Li Q, Dai Z, Ma J, Zhou H, Li G, Meng C, Chen Y. Arginine methylation of PPP1CA by CARM1 regulates glucose metabolism and affects osteogenic differentiation and osteoclastic differentiation. Clin Transl Med 2023; 13:e1369. [PMID: 37649137 PMCID: PMC10468565 DOI: 10.1002/ctm2.1369] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND The imbalance between osteoblasts and osteoclasts may lead to osteoporosis. Osteoblasts and osteoclasts have different energy requirements, with aerobic glycolysis being the prominent metabolic feature of osteoblasts, while osteoclast differentiation and fusion are driven by oxidative phosphorylation. METHODS By polymerase chain reaction as well as Western blotting, we assayed coactivator-associated arginine methyltransferase 1 (CARM1) expression in bone tissue, the mouse precranial osteoblast cell line MC3T3-E1 and the mouse monocyte macrophage leukaemia cell line RAW264.7, and expression of related genes during osteogenic differentiation and osteoclast differentiation. Using gene overexpression (lentivirus) and loss-of-function approach (CRISPR/Cas9-mediated knockout) in vitro, we examined whether CARM1 regulates osteogenic differentiation and osteoblast differentiation by metabolic regulation. Transcriptomic assays and metabolomic assays were used to find the mechanism of action of CARM1. Furthermore, in vitro methylation assays were applied to clarify the arginine methylation site of PPP1CA by CARM1. RESULTS We discovered that CARM1 reprogrammed glucose metabolism in osteoblasts and osteoclasts from oxidative phosphorylation to aerobic glycolysis, thereby promoting osteogenic differentiation and inhibiting osteoclastic differentiation. In vivo experiments revealed that CARM1 significantly decreased bone loss in osteoporosis model mice. Mechanistically, CARM1 methylated R23 of PPP1CA, affected the dephosphorylation of AKT-T450 and AMPK-T172, and increased the activities of phosphofructokinase-1 and pructose-2,6-biphosphatase3, causing an up-regulation of glycolytic flux. At the same time, as a transcriptional coactivator, CARM1 regulated the expression of pyruvate dehydrogenase kinase 3, which resulted in the inhibition of pyruvate dehydrogenase activity and inhibition of the tricarboxylic acid cycle, leading to a subsequent decrease in the flux of oxidative phosphorylation. CONCLUSIONS These findings reveal for the first time the mechanism by which CARM1 affects both osteogenesis and osteoclast differentiation through metabolic regulation, which may represent a new feasible treatment strategy for osteoporosis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of MicroorthopaedicsAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanShandongChina
- Department of Spine SurgeryAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Guangjun Jiao
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yunhao You
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Xiang Li
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Jincheng Liu
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Zhenqian Sun
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Qinghui Li
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Zihan Dai
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Jinlong Ma
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
- Department of OrthopaedicsThe First Clinical College of Shandong UniversityJinanShandongChina
| | - Hongming Zhou
- Department of Spine SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Department of Spine SurgeryLinyi Central HospitalLinyiShandongChina
| | - Gang Li
- Department of MicroorthopaedicsAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanShandongChina
| | - Chunyang Meng
- Department of Spine SurgeryAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Yunzhen Chen
- Department of Spine SurgeryQilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
3
|
You Y, Liu J, Zhang L, Li X, Sun Z, Dai Z, Ma J, Jiao G, Chen Y. WTAP-mediated m 6A modification modulates bone marrow mesenchymal stem cells differentiation potential and osteoporosis. Cell Death Dis 2023; 14:33. [PMID: 36650131 PMCID: PMC9845239 DOI: 10.1038/s41419-023-05565-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
An imbalance in the differentiation potential of bone marrow mesenchymal stem cells (BMSCs) is an important pathogenic mechanism underlying osteoporosis (OP). N6-methyladenosine (m6A) is the most common post-transcriptional modification in eukaryotic cells. The role of the Wilms' tumor 1-associated protein (WTAP), a member of the m6A functional protein family, in regulating BMSCs differentiation remains unknown. We used patient-derived and mouse model-derived samples, qRT-PCR, western blot assays, ALP activity assay, ALP, and Alizarin Red staining to determine the changes in mRNA and protein levels of genes and proteins associated with BMSCs differentiation. Histological analysis and micro-CT were used to evaluate developmental changes in the bone. The results determined that WTAP promoted osteogenic differentiation and inhibited adipogenic differentiation of BMSCs. We used co-immunoprecipitation (co-IP), RNA immunoprecipitation (RIP), methylated RNA immunoprecipitation (MeRIP), RNA pulldown, and dual-luciferase assay to explore the direct mechanism. Mechanistically, the expression of WTAP increased during osteogenic differentiation and significantly promoted pri-miR-181a and pri-miR-181c methylation, which was recognized by YTHDC1, and increased the maturation to miR-181a and miR-181c. MiR-181a and miR-181c inhibited the mRNA expression of SFRP1, promoting the osteogenic differentiation of BMSCs. Our results demonstrated that the WTAP/YTHDC1/miR-181a and miR-181c/SFRP1 axis regulated the differentiation fate of BMSCs, suggesting that it might be a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Yunhao You
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Jincheng Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Lu Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Xiang Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Zhenqian Sun
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Zihan Dai
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Jinlong Ma
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Guangjun Jiao
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China
| | - Yunzhen Chen
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012, Jinan, Shandong, China.
| |
Collapse
|
4
|
Behera J, Ison J, Voor MJ, Tyagi N. Exercise-Linked Skeletal Irisin Ameliorates Diabetes-Associated Osteoporosis by Inhibiting the Oxidative Damage-Dependent miR-150-FNDC5/Pyroptosis Axis. Diabetes 2022; 71:2777-2792. [PMID: 35802043 PMCID: PMC9750954 DOI: 10.2337/db21-0573] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/22/2022] [Indexed: 01/25/2023]
Abstract
Recent evidence suggests that physical exercise (EX) promotes skeletal development. However, the impact of EX on the progression of bone loss and deterioration of mechanical strength in mice with type 2 diabetic mellitus (T2DM) remains unexplored. In the current study, we investigated the effect of EX on bone mass and mechanical quality using a diabetic mouse model. The T2DM mouse model was established with a high-fat diet with two streptozotocin injections (50 mg/kg/body wt) in C57BL/6 female mice. The diabetic mice underwent treadmill exercises (5 days/week at 7-11 m/min for 60 min/day) for 8 weeks. The data showed that diabetes upregulated miR-150 expression through oxidative stress and suppressed FNDC5/Irisin by binding to its 3'-untranslated region. The decreased level of irisin further triggers the pyroptosis response in diabetic bone tissue. EX or N-acetyl cysteine or anti-miRNA-150 transfection in T2DM mice restored FNDC5/Irisin expression and bone formation. Furthermore, EX or recombinant irisin administration prevented T2DM-Induced hyperglycemia and improved glucose intolerance in diabetic mice. Furthermore, osteoblastic knockdown of Nlrp3 silencing (si-Nlrp3) or pyroptosis inhibitor (Ac-YVADCMK [AYC]) treatment restores bone mineralization in diabetic mice. Micro-computed tomography scans and mechanical testing revealed that trabecular bone microarchitecture and bone mechanical properties were improved after EX in diabetic mice. Irisin, either induced by skeleton or daily EX or directly administered, prevents bone loss by mitigating inflammasome-associated pyroptosis signaling in diabetic mice. This study demonstrates that EX-induced skeletal irisin ameliorates diabetes-associated glucose intolerance and bone loss and possibly provides a mechanism of its effects on metabolic osteoporosis.
Collapse
Affiliation(s)
- Jyotirmaya Behera
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY
| | - Jessica Ison
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY
| | - Michael J. Voor
- Departments of Orthopaedic Surgery and Bioengineering, School of Medicine, University of Louisville, Louisville, KY
- Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY
| | - Neetu Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY
| |
Collapse
|
5
|
Tanjaya J, Ha P, Zhang Y, Wang C, Shah Y, Berthiaume E, Pan HC, Shi J, Kwak J, Wu B, Ting K, Zhang X, Soo C. Genetic and pharmacologic suppression of PPARγ enhances NELL-1-stimulated bone regeneration. Biomaterials 2022; 287:121609. [PMID: 35839586 PMCID: PMC10434299 DOI: 10.1016/j.biomaterials.2022.121609] [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: 05/16/2021] [Revised: 05/15/2022] [Accepted: 05/28/2022] [Indexed: 11/02/2022]
Abstract
Recent investigations into mechanisms behind the development of osteoporosis suggest that suppressing PPARγ-mediated adipogenesis can improve bone formation and bone mineral density. In this study, we investigated a co-treatment strategy to enhance bone formation by combining NELL-1, an osteogenic molecule that has been extensively studied for its potential use as a therapeutic for osteoporosis, with two methods of PPARγ suppression. First, we suppressed PPARγ genetically using lentiviral PPARγ-shRNA in immunocompromised mice for a proof of concept. Second, we used a PPARγ antagonist to suppress PPARγ pharmacologically in immunocompetent senile osteopenic mice for clinical transability. We found that the co-treatment strategy significantly increased bone formation, increased the proliferation stage cell population, decreased late apoptosis of primary mouse BMSCs, and increased osteogenic marker mRNA levels in comparison to the single agent treatment groups. The addition of PPARγ suppression to NELL-1 therapy enhanced NELL-1's effects on bone formation by upregulating anabolic processes without altering NELL-1's inhibitory effects on osteoclastic and adipogenic activities. Our findings suggest that combining PPARγ suppression with therapeutic NELL-1 may be a viable method that can be further developed as a novel strategy to reverse bone loss and decrease marrow adiposity in age-related osteoporosis.
Collapse
Affiliation(s)
- Justine Tanjaya
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Pin Ha
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Yulong Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, USA, 90025; Weintraub Center for Reconstructive Biotechnology, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Chenchao Wang
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Yash Shah
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Emily Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Hsin Chuan Pan
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Jiayu Shi
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Jinny Kwak
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Benjamin Wu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, USA, 90025; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA, 90025; Weintraub Center for Reconstructive Biotechnology, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Kang Ting
- Forsyth Institute, Harvard University, Cambridge, MA, USA, 02142.
| | - Xinli Zhang
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, USA, 90025; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA, 90025; Weintraub Center for Reconstructive Biotechnology, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025.
| |
Collapse
|
6
|
Wang H, Shi Y, He F, Ye T, Yu S, Miao H, Liu Q, Zhang M. GDF11 inhibits abnormal adipogenesis of condylar chondrocytes in temporomandibular joint osteoarthritis. Bone Joint Res 2022; 11:453-464. [PMID: 35787089 PMCID: PMC9350697 DOI: 10.1302/2046-3758.117.bjr-2022-0019.r1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aims Abnormal lipid metabolism is involved in the development of osteoarthritis (OA). Growth differentiation factor 11 (GDF11) is crucial in inhibiting the differentiation of bone marrow mesenchymal stem cells into adipocytes. However, whether GDF11 participates in the abnormal adipogenesis of chondrocytes in OA cartilage is still unclear. Methods Six-week-old female mice were subjected to unilateral anterior crossbite (UAC) to induce OA in the temporomandibular joint (TMJ). Histochemical staining, immunohistochemical staining (IHC), and quantitative real-time polymerase chain reaction (qRT-PCR) were performed. Primary condylar chondrocytes of rats were stimulated with fluid flow shear stress (FFSS) and collected for oil red staining, immunofluorescence staining, qRT-PCR, and immunoprecipitation analysis. Results Abnormal adipogenesis, characterized by increased expression of CCAAT/enhancer-binding protein α (CEBPα), fatty acid binding protein 4 (FABP4), Perilipin1, Adiponectin (AdipoQ), and peroxisome proliferator-activated receptor γ (PPARγ), was enhanced in the degenerative cartilage of TMJ OA in UAC mice, accompanied by decreased expression of GDF11. After FFSS stimulation, there were fat droplets in the cytoplasm of cultured cells with increased expression of PPARγ, CEBPα, FABP4, Perilipin1, and AdipoQ and decreased expression of GDF11. Exogenous GDF11 inhibited increased lipid droplets and expression of AdipoQ, CEBPα, and FABP4 induced by FFSS stimulation. GDF11 did not affect the change in PPARγ expression under FFSS, but promoted its post-translational modification by small ubiquitin-related modifier (SUMOylation). Local injection of GDF11 alleviated TMJ OA-related cartilage degeneration and abnormal adipogenesis in UAC mice. Conclusion Abnormal adipogenesis of chondrocytes and decreased GDF11 expression were observed in degenerative cartilage of TMJ OA. GDF11 supplementation effectively inhibits the adipogenesis of chondrocytes and thus alleviates TMJ condylar cartilage degeneration. GDF11 may inhibit the abnormal adipogenesis of chondrocytes by affecting the SUMOylation of PPARγ. Cite this article: Bone Joint Res 2022;11(7):453–464.
Collapse
Affiliation(s)
- Helin Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases, Department of Medical Rehabilitation, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yuqian Shi
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Feng He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Tao Ye
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Shibin Yu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Hui Miao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Qian Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Mian Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
7
|
Wang C, Tanjaya J, Shen J, Lee S, Bisht B, Pan HC, Pang S, Zhang Y, Berthiaume EA, Chen E, Da Lio AL, Zhang X, Ting K, Guo S, Soo C. Peroxisome Proliferator-Activated Receptor-γ Knockdown Impairs Bone Morphogenetic Protein-2-Induced Critical-Size Bone Defect Repair. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:648-664. [PMID: 30593824 PMCID: PMC6412314 DOI: 10.1016/j.ajpath.2018.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/13/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
The Food and Drug Administration-approved clinical dose (1.5 mg/mL) of bone morphogenetic protein-2 (BMP2) has been reported to induce significant adverse effects, including cyst-like adipose-infiltrated abnormal bone formation. These undesirable complications occur because of increased adipogenesis, at the expense of osteogenesis, through BMP2-mediated increases in the master regulatory gene for adipogenesis, peroxisome proliferator-activated receptor-γ (PPARγ). Inhibiting PPARγ during osteogenesis has been suggested to drive the differentiation of bone marrow stromal/stem cells toward an osteogenic, rather than an adipogenic, lineage. We demonstrate that knocking down PPARγ while concurrently administering BMP2 can reduce adipogenesis, but we found that it also impairs BMP2-induced osteogenesis and leads to bone nonunion in a mouse femoral segmental defect model. In addition, in vitro studies using the mouse bone marrow stromal cell line M2-10B4 and mouse primary bone marrow stromal cells confirmed that PPARγ knockdown inhibits BMP2-induced adipogenesis; attenuates BMP2-induced cell proliferation, migration, invasion, and osteogenesis; and escalates BMP2-induced cell apoptosis. More important, BMP receptor 2 and 1B expression was also significantly inhibited by the combined BMP2 and PPARγ knockdown treatment. These findings indicate that PPARγ is critical for BMP2-mediated osteogenesis during bone repair. Thus, uncoupling BMP2-mediated osteogenesis and adipogenesis using PPARγ inhibition to combat BMP2's adverse effects may not be feasible.
Collapse
Affiliation(s)
- Chenchao Wang
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, People's Republic of China; Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Justine Tanjaya
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Jia Shen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Soonchul Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Bharti Bisht
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Hsin Chuan Pan
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Shen Pang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Yulong Zhang
- Departments of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, California
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Eric Chen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Andrew L Da Lio
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Shu Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, People's Republic of China.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
8
|
Zhang L, Chang L, Xu J, Meyers CA, Yan N, Zou E, Ding C, Ting K, Soo C, Pang S, James AW. Frontal Bone Healing Is Sensitive to Wnt Signaling Inhibition via Lentiviral-Encoded Beta-Catenin Short Hairpin RNA. Tissue Eng Part A 2018; 24:1742-1752. [PMID: 29929440 PMCID: PMC6302677 DOI: 10.1089/ten.tea.2017.0465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/24/2018] [Indexed: 01/10/2023] Open
Abstract
The Wnt/β-catenin signaling pathway plays an integral role in skeletal biology, spanning from embryonic skeletal patterning through bone maintenance and bone repair. Most experimental methods to antagonize Wnt signaling in vivo are either systemic or transient, including genetic approaches, use of small-molecule inhibitors, or neutralizing antibodies. We sought to develop a novel, localized model of prolonged Wnt/β-catenin signaling blockade by the application and validation of a lentivirus encoding β-catenin short hairpin RNA (shRNA). Efficacy of lentiviral-encoded β-catenin shRNA was first confirmed in vitro using bone marrow mesenchymal stromal cells, and in vivo using an intramedullary long bone injection model in NOD SCID mice. Next, the effects of β-catenin knockdown were assessed in a calvarial bone defect model, in which the frontal bone demonstrates enhanced bone healing associated with heightened Wnt/β-catenin signaling. Lentivirus encoding either β-catenin shRNA or random sequence shRNA with enhanced green fluorescent protein (control) was injected overlying the calvaria of NOD SCID mice and bone defects were created in either the frontal or parietal bones. Among mice treated with lentivirus encoding β-catenin shRNA, frontal bone defect healing was significantly reduced by all radiographic and histologic metrics. In contrast, parietal bone healing was minimally impacted by β-catenin shRNA. In aggregate, our data document the application and validation of a lentivirus encoding β-catenin shRNA model that represents an easily replicable tool for examining the importance of locoregional Wnt/β-catenin signaling in bone biology and regeneration.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, China
| | - Leslie Chang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
- University of California San Diego School of Medicine, La Jolla, California
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | | | - Noah Yan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Erin Zou
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Catherine Ding
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| | - Kang Ting
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Shen Pang
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| |
Collapse
|
9
|
Grol MW, Stone A, Ruan MZ, Guse K, Lee BH. Prospects of Gene Therapy for Skeletal Diseases. GENETICS OF BONE BIOLOGY AND SKELETAL DISEASE 2018:119-137. [DOI: 10.1016/b978-0-12-804182-6.00008-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
10
|
Li G, Xu Z, Hou L, Li X, Li X, Yuan W, Polat M, Chang S. Differential effects of bisphenol A diglicydyl ether on bone quality and marrow adiposity in ovary-intact and ovariectomized rats. Am J Physiol Endocrinol Metab 2016; 311:E922-E927. [PMID: 27756728 DOI: 10.1152/ajpendo.00267.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 11/22/2022]
Abstract
Bisphenol A diglycidyl ether (BADGE), a PPARγ2 antagonist, has been shown to inhibit marrow adipogenesis and promote bone formation in intact animals. We investigated the impact of BADGE on a new and more clinically relevant physiological model, the ovariectomized (OVX) rat model. Forty female Wistar rats were divided into four treatment groups for 12 wk (n = 10/group): sham+vehicle, sham+BADGE, OVX+vehicle, and OVX+BADGE. Postmortem analyses included MRI, micro-CT, serological test, histomorphometry, biomechanical tests, RT-PCR, and Western blot. Overall, OVX induced a sequential marrow fat expansion accompanied by bone deterioration. Compared with OVX controls, BADGE reduced fat fraction of the distal femur by 36.3%, adipocyte density by 33.0%, adipocyte size by 28.6%, adipocyte volume percentage by 57.8%, and adipogenic markers PPARγ2 and C/EBPα by ∼50% in OVX rats. Similar results were observed in sham rats vs. vehicle. BADGE could promote bone quality in sham rats; however, BADGE did not significantly improve trabecular microarchitecture, biomechanical strength, and dynamic histomorphometric parameters except for trabecular separation in OVX rats. We concluded that early BADGE treatment at a dose of 30 mg/kg attenuates marrow adiposity in ovary-intact and OVX rats and stimulates bone formation in ovary-intact rats but does not significantly rescue bone quality in OVX rats.
Collapse
Affiliation(s)
- Guanwu Li
- Department of Radiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Zheng Xu
- Xinzhuang Community Health Center, Shanghai, China
| | - Lingmi Hou
- Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Xuefeng Li
- Department of Radiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Li
- Department of Gerontology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Yuan
- Department of Spinal Disease Unit, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; and
| | - Maki Polat
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Shixin Chang
- Department of Radiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
11
|
Chung JE, Park JH, Yun JW, Kang YH, Park BW, Hwang SC, Cho YC, Sung IY, Woo DK, Byun JH. Cultured Human Periosteum-Derived Cells Can Differentiate into Osteoblasts in a Perioxisome Proliferator-Activated Receptor Gamma-Mediated Fashion via Bone Morphogenetic Protein signaling. Int J Med Sci 2016; 13:806-818. [PMID: 27877072 PMCID: PMC5118751 DOI: 10.7150/ijms.16484] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/13/2016] [Indexed: 01/02/2023] Open
Abstract
The differentiation of mesenchymal stem cells towards an osteoblastic fate depends on numerous signaling pathways, including activation of bone morphogenetic protein (BMP) signaling components. Commitment to osteogenesis is associated with activation of osteoblast-related signal transduction, whereas inactivation of this signal transduction favors adipogenesis. BMP signaling also has a critical role in the processes by which mesenchymal stem cells undergo commitment to the adipocyte lineage. In our previous study, we demonstrated that an agonist of the perioxisome proliferator-activated receptor γ (PPARγ), a master regulator of adipocyte differentiation, stimulates osteoblastic differentiation of cultured human periosteum-derived cells. In this study, we used dorsomorphin, a selective small molecule inhibitor of BMP signaling, to investigate whether BMP signaling is involved in the positive effects of PPARγ agonists on osteogenic phenotypes of cultured human periosteum-derived cells. Both histochemical detection and bioactivity of ALP were clearly increased in the periosteum-derived cells treated with the PPARγ agonist at day 10 of culture. Treatment with the PPARγ agonist also caused an increase in alizarin red S staining and calcium content in the periosteum-derived osteoblasts at 2 and 3 weeks of culture. In contrast, dorsomorphin markedly decreased ALP activity, alizarin red S staining and calcium content in both the cells treated with PPARγ agonist and the cells cultured in osteogenic induction media without PPARγ agonist during the culture period. In addition, the PPARγ agonist clearly increased osteogenic differentiation medium-induced BMP-2 upregulation in the periosteum-derived osteoblastic cells at 2 weeks of culture as determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR), immunoblotting, and immunocytochemical analyses. Although further study will be needed to clarify the mechanisms of PPARγ-regulated osteogenesis, our results suggest that the positive effects of a PPARγ agonist on the osteogenic phenotypes of cultured human periosteum-derived cells seem to be dependent on BMP signaling.
Collapse
Affiliation(s)
- Jin-Eun Chung
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 660-702, Republic of Korea
| | - Jin-Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 660-702, Republic of Korea
| | - Jeong-Won Yun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 660-702, Republic of Korea
| | - Young-Hoon Kang
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 660-702, Republic of Korea
| | - Bong-Wook Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 660-702, Republic of Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Yeong-Cheol Cho
- Department of Oral and Maxillofacial Surgery, College of Medicine, Ulsan University Hospital, University of Ulsan, Ulsan, Republic of Korea
| | - Iel-Yong Sung
- Department of Oral and Maxillofacial Surgery, College of Medicine, Ulsan University Hospital, University of Ulsan, Ulsan, Republic of Korea
| | - Dong Kyun Woo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 660-702, Republic of Korea
| |
Collapse
|
12
|
Wang Y, Zhu G, Wang J, Chen J. Irradiation alters the differentiation potential of bone marrow mesenchymal stem cells. Mol Med Rep 2015; 13:213-23. [PMID: 26572960 PMCID: PMC4686093 DOI: 10.3892/mmr.2015.4539] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022] Open
Abstract
Bone injury following radiotherapy has been confirmed by epidemiological and animal studies. However, the underlying mechanism remains to be elucidated and no preventive or curative solution has been identified for this bone loss. The present study aimed to investigate the irradiation‑altered osteogenesis and adipogenesis of bone marrow mesenchymal stem cells (BMSCs). BMSCs were derived and exposed to γ‑irradiation at doses of 0, 0.25, 0.5, 1, 2, 5 and 10 Gy. Cell viability was assessed using a 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5 diphenyl tetrazolium bromide assay, and clonal expansion in vitro was detected by colony forming unit assessment. The osteogenic differentiation ability was demonstrated by alkaline phosphatase (ALP) activity, ALP staining and mineralization alizarin red staining, and the adipogenic differentiation ability was determined using Oil O red staining. The osteogenesis‑associated genes, RUNX2, ALP, osteocalcin (OCN) and adipogenesis‑associated genes, PPAR‑γ and C/EBPα, were detected using reverse transcription‑quantitative polymerase chain reaction analyses. The protein expression levels of RUNX2, ALP and PPAR‑γ were detected using western blotting. Compared with the control, significant decreases in the proliferation, ALP activity and mineralization ability of the BMSCs were observed in the γ‑irradiation group, with a high level of correlation with the exposure dose. However, no significant changes were observed in the area of Oil red O positive staining. The mRNA levels of RUNX2, ALP and OCN were decreased (P<0.05), however, no significant changes were observed in the levels of C/EBPα and PPAR‑γ. The protein expression levels of RUNX2 and ALP were decreased in the irradiated BMSCs, however, no significant difference was observed in the protein expression of PPAR‑γ. Irradiation inhibited the osteogenic and adipogenic ability of the BMSCs, and the osteogenic differentiation was decreased. The results of the present study provided evidence to assist in further elucidating radiotherapy‑associated side effects on the skeleton.
Collapse
Affiliation(s)
- Yu Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Guoying Zhu
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jianping Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Junxiang Chen
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
13
|
Shao M, Liu C, Song Y, Ye W, He W, Yuan G, Gu S, Lin C, Ma L, Zhang Y, Tian W, Hu T, Chen Y. FGF8 signaling sustains progenitor status and multipotency of cranial neural crest-derived mesenchymal cells in vivo and in vitro. J Mol Cell Biol 2015; 7:441-454. [PMID: 26243590 PMCID: PMC4589951 DOI: 10.1093/jmcb/mjv052] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 04/19/2015] [Accepted: 04/27/2015] [Indexed: 02/05/2023] Open
Abstract
The cranial neural crest (CNC) cells play a vital role in craniofacial development and regeneration. They are multi-potent progenitors, being able to differentiate into various types of tissues. Both pre-migratory and post-migratory CNC cells are plastic, taking on diverse fates by responding to different inductive signals. However, what sustains the multipotency of CNC cells and derivatives remains largely unknown. In this study, we present evidence that FGF8 signaling is able to sustain progenitor status and multipotency of CNC-derived mesenchymal cells both in vivo and in vitro. We show that augmented FGF8 signaling in pre-migratory CNC cells prevents cell differentiation and organogenesis in the craniofacial region by maintaining their progenitor status. CNC-derived mesenchymal cells with Fgf8 overexpression or control cells in the presence of exogenous FGF8 exhibit prolonged survival, proliferation, and multi-potent differentiation capability in cell cultures. Remarkably, exogenous FGF8 also sustains the capability of CNC-derived mesenchymal cells to participate in organogenesis such as odontogenesis. Furthermore, FGF8-mediated signaling strongly promotes adipogenesis but inhibits osteogenesis of CNC-derived mesenchymal cells in vitro. Our results reveal a specific role for FGF8 in the maintenance of progenitor status and in fate determination of CNC cells, implicating a potential application in expansion and fate manipulation of CNC-derived cells in stem cell-based craniofacial regeneration.
Collapse
Affiliation(s)
- Meiying Shao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Chao Liu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Yingnan Song
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA Southern Center for Biomedical Research, Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou 350108, China
| | - Wenduo Ye
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Wei He
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Guohua Yuan
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA Hubei-MOST KLOS and KLOBM School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Shuping Gu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Congxin Lin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Liang Ma
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research, Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou 350108, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA Southern Center for Biomedical Research, Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou 350108, China
| |
Collapse
|
14
|
Abstract
The night and day cycle governs the circadian (24 hourly) rhythm of activity and rest in animals and humans. This is reflected in daily changes of the global gene expression pattern and metabolism, but also in the local physiology of various tissues. A central clock in the brain co-ordinates the rhythmic locomotion behaviour, as well as synchronizing various local oscillators, such as those found in the musculoskeletal system. It has become increasingly recognized that the internal molecular clocks in cells allow a tissue to anticipate the rhythmic changes in their local environment and the specific demands of that tissue. Consequently, the majority of the rhythmic clock controlled genes and pathways are tissue specific. The concept of the tissue-specific function of circadian clocks is further supported by the diverse musculoskeletal phenotypes in mice with deletions or mutations of various core clock components, ranging from increased bone mass, dwarfism, arthropathy, reduced muscle strength and tendon calcification. The present review summarizes the current understanding of the circadian clocks in muscle, bone, cartilage and tendon tissues, with particular focus on the evidence of circadian rhythms in tissue physiology, their entrainment mechanisms and disease links, and the tissue-specific clock target genes/pathways. Research in this area holds strong potential to advance our understanding of how circadian rhythms control the health and disease of the musculoskeletal tissues, which has major implications in diseases associated with advancing age. It could also have potential implications in sports performance and sports medicine.
Collapse
|