1
|
Shen J, Pei Y, Bai S, Lei S, Xia S, Zhang J, Li X, Xu H, Zheng X, Shen X, Zhao H, Liu L, Yang X, Wang X. Magnesium-based implants accelerate femoral fracture healing through promoting histone lactylation-mediated osteoclastogenesis inhibition. Life Sci 2025:123639. [PMID: 40252757 DOI: 10.1016/j.lfs.2025.123639] [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: 01/22/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
AIMS To investigate the molecular mechanisms by which magnesium (Mg)-based implants, specifically Mg-containing intramedullary nails (Mg-IMNs), promote femoral fracture healing. MATERIALS AND METHODS Rats with femoral fractures were treated with Mg-IMNs. In vitro experiments were conducted to assess the impact of Mg2+ on osteoclastogenesis and histone lactylation. Histological analysis, Western blotting, and qRT-PCR were employed to evaluate osteoclast maturation and the molecular pathways involved. In vivo, lactate was administered to replicate Mg-IMN effects, and lactate production was inhibited to observe potential reversal effects. KEY FINDINGS Mg-IMNs significantly enhanced fracture healing by inhibiting osteoclastogenesis. Mg2+ promoted intracellular lactate production, leading to histone lactylation, which suppressed osteoclast maturation by downregulating NFATc1. The P300/H3K18LA/HDAC1 pathway was identified as a key mediator in this process. Additionally, lactate administration mimicked the effects of Mg-IMNs, while blocking lactate reversed these effects. SIGNIFICANCE This study uncovers a novel mechanism by which Mg2+ promotes fracture healing through histone lactylation-mediated inhibition of osteoclastogenesis. These findings offer new therapeutic strategies for enhancing fracture repair via epigenetic regulation.
Collapse
Affiliation(s)
- Junyi Shen
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Shangying Bai
- China-America Institute of Neuroscience, Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Simeng Lei
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Suhang Xia
- Department of Joint Diseases, Zhengzhou Orthopaedics Hospital, Zhengzhou, Henan, China
| | - Jie Zhang
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xingyu Li
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Hanchi Xu
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xinyu Zheng
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xuezhen Shen
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Huanjun Zhao
- Department of Burn Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liang Liu
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Xinlin Yang
- Orthopaedic Research Lab, University of Virginia, Charlottesville, VA, USA.
| | - Xuefei Wang
- Department of Orthopaedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Jiang Y, Zhang D, He X, Chen C, Xie L, Liu L, Yu Z, Zhang Y, Zheng J, Huang D. BCAT1 contributes to the development of TKI-resistant CML. Cell Oncol (Dordr) 2025; 48:411-424. [PMID: 39412615 PMCID: PMC11996995 DOI: 10.1007/s13402-024-01003-y] [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] [Accepted: 10/03/2024] [Indexed: 04/15/2025] Open
Abstract
PURPOSE Although most of chronic myeloid leukemia (CML) patients can be effectively treated by the tyrosine kinase inhibitors (TKIs), such as Imatinib, TKI-resistance still occurs in approximately 15-17% of cases. Although many studies indicate that branched chain amino acid (BCAA) metabolism may contribute to the TKI resistance in CML, the detailed mechanisms remains largely unknown. METHOD The cell proliferation, colony formation and in vivo transplantation were used to determined the functions of BCAT1 in leukemogenesis. Quantitative real-time PCR (RT-PCR), western blotting, RNA sequencing, BCAA stimulation in vitro were applied to characterize the underlying molecular mechanism that control the leukemogenic activity of BCAT1-knockdown cells. RESULTS In this report, we revealed that branched chain amino acid transaminase 1 (BCAT1) is highly enriched in both mouse and human TKI-resistant CML cells. Leukemia was almost completely abrogated upon BCAT1 knockdown during transplantation in a BCR-ABLT315I-induced murine TKI-resistant CML model. Moreover, knockdown of BCAT1 led to a dramatic decrease in the proliferation of TKI-resistant human leukemia cell lines. BCAA/BCAT1 signaling enhanced the phosphorylation of CREB, which is required for maintenance of TKI-resistant CML cells. Importantly, blockade of BCAA/BCAT1 signaling efficiently inhibited leukemogenesis both in vivo and in vitro. CONCLUSIONS These findings demonstrate the role of BCAA/BCAT1 signaling in cancer development and suggest that targeting BCAA/BCAT1 signaling is a potential strategy for interfering with TKI-resistant CML.
Collapse
MESH Headings
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Humans
- Animals
- Cell Proliferation/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Transaminases/metabolism
- Transaminases/genetics
- Cell Line, Tumor
- Mice
- Gene Knockdown Techniques
- Imatinib Mesylate/pharmacology
Collapse
Affiliation(s)
- Yu Jiang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Difan Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaoxiao He
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Chiqi Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Li Xie
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Ligen Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Zhuo Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yaping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dan Huang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
3
|
Wang H, Guan Y, Lin L, Qiang Z, Huo Y, Zhu L, Yan B, Shao S, Liu W, Yang J. DEC1 deficiency promotes osteoclastic activity by augmenting NFATc1 signaling via transactivation and the Ca 2+/calcineurin pathway. Biochem Pharmacol 2025; 233:116754. [PMID: 39824467 DOI: 10.1016/j.bcp.2025.116754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/30/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
We have previously demonstrated that DEC1 promotes osteoblast differentiation. This study aims to evaluate the impact of DEC1 knockout on osteopenic activities, such as osteoclast differentiation and the expression of bone-degrading genes. To gain mechanistic insights, we employed both in vivo and in vitro experiments, utilizing cellular and molecular approaches, including osteoclast differentiation assays and RNA-seq in combination with ChIP-seq. Our results showed that NFATc1, a master regulator of osteoclast differentiation, and PPP3CB, a member of the calcineurin family, were significantly upregulated in DEC1-/- mice. In vitro experiments revealed that osteoclast differentiation significantly increased both the number and size of osteoclasts in DEC1-/- bone marrow macrophages (BMMs) compared to DEC1+/+ BMMs. Additionally, NFATc1 expression was notably higher in DEC1-/- BMMs than in DEC1+/+ BMMs. Overexpression of DEC1 reduced NFATc1 promoter activity, while knockout increased it. Furthermore, intracellular free Ca2+ levels and calcineurin activity were elevated (∼150 %) in DEC1-/- BMMs compared to DEC1+/+ BMMs. Importantly, the use of calcineurin inhibitors and calcium channel blockers effectively abolished the increased osteoclast differentiation observed in DEC1-/- BMMs. In summary, DEC1 deficiency promotes osteoclast differentiation by enhancing NFATc1 signaling through transcriptional regulation and the Ca2+/calcineurin pathway. Clinically, the mRNA levels of DEC1 were reduced by up to 75 % in patients with osteoporosis. The findings of this study establish that inducing DEC1 expression, alongside attenuators of the Ca2+/calcineurin pathway, offers a molecular basis for preventing and treating osteoporosis associated with DEC1 deficiency.
Collapse
Affiliation(s)
- Haobin Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Yu Guan
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Lan Lin
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Zhiyi Qiang
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Ying Huo
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Ling Zhu
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Bingfang Yan
- James L. Winkle College of Pharmacy University of Cincinnati, Cincinnati, OH 45229, USA
| | - Shulin Shao
- Nanjing Pukou District Traditional Chinese Medicine Hospital, Nanjing 210000, PR China
| | - Wei Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China
| | - Jian Yang
- Department of Pharmacology, Nanjing Medical University, Nanjing 211166, PR China.
| |
Collapse
|
4
|
Jiang S, Wang Y, Ren Y, Tang Q, Xue C, Wang Z, Zhang Q, Hu Y, Wang H, Zhao F, Zhu MX, Cao Z. TRPC6 suppresses liver fibrosis by inhibiting hepatic stellate cell activation via CaMK4-CREB pathway. Br J Pharmacol 2025. [PMID: 39887689 DOI: 10.1111/bph.17431] [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: 05/16/2024] [Revised: 09/26/2024] [Accepted: 12/06/2024] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND AND PURPOSE Genetic ablation or inhibition of the cation channel TRPC6 is protective against renal, cardiac and intestinal fibrosis. However, TRPC6 expression is decreased in patients with liver diseases. Here, we explored the role of TRPC6 in liver fibrosis and the underlying mechanism. EXPERIMENTAL APPROACH Bile duct ligation and thioacetamide gavage were used to model liver fibrosis in C57BL/6J mice. Western blotting, immunolabelling and qPCR were employed for protein and mRNA expression. Liver injury/fibrosis were assessed using serum alanine transaminase and aspartate transaminase assays, haematoxylin-eosin, Masson and Sirius red staining. Adenoviruses were used to overexpress TRPC6 and CREB1Y134F. ChIP and dual-luciferase reporter assays were performed to test the direct inhibition of Acta2 transcription by CREB. KEY RESULTS TRPC6 protein levels were decreased in fibrotic liver tissues from both patients and mice, with the decrease being more robust in fibrotic areas. In hepatic stellate cells (HSCs), TRPC6 ablation aggravated liver injury and fibrosis, which was alleviated by overexpressing TRPC6. In primary cultured HSCs, deletion of TRPC6 exacerbated self-activation of HSCs, which was reversed by restoration of TRPC6 expression. Mechanistically, TRPC6 suppressed HSC activation through CaMK4-mediated CREB phosphorylation. CREB directly interacted with the promoter region of Acta2 to inhibit its transcription. Expression of a constitutively active form of CREB1 (CREB1Y134F) in HSCs attenuated BDL-induced liver injury/fibrosis in TRPC6 knockout mice. CONCLUSION AND IMPLICATIONS Deficiency of TRPC6 aggravates liver injury/fibrosis through augmentation of HSC activation. Increasing TRPC6 expression/function would be therapeutically beneficial for fibrotic liver diseases.
Collapse
Affiliation(s)
- Shan Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yujing Wang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Younan Ren
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qinglian Tang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chu Xue
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhi Wang
- Department of Gastroenterology, Zhongda Hospital, Nanjing, China
| | - Qi Zhang
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yixin Hu
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai, China
| | - Fang Zhao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Ray S, McCall JL, Tian JB, Jeon J, Douglas A, Tyler K, Liu S, Berry K, Nicewarner B, Hall C, Groschner K, Bacsa B, Geldenhuys W, Zhu MX, Blair HC, Barnett JB, Soboloff J. Targeting TRPC channels for control of arthritis-induced bone erosion. SCIENCE ADVANCES 2025; 11:eabm9843. [PMID: 39813349 PMCID: PMC11734723 DOI: 10.1126/sciadv.abm9843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025]
Abstract
Arthritis leads to bone erosion due to an imbalance between osteoclast and osteoblast function. Our prior investigations revealed that the Ca2+-selective ion channel, Orai1, is critical for osteoclast maturation. Here, we show that the small-molecule ELP-004 preferentially inhibits transient receptor potential canonical (TRPC) channels. While ELP-004 minimally affected physiological RANKL-induced osteoclast maturation in murine bone marrow- and spleen-derived myeloid cells (BMSMCs) and human PBMC-derived cells, it potently interfered with osteoclast maturation driven by TNFα or LTB4. The contribution of TRPC channels to osteoclastogenesis was examined using BMSMCs derived from TRPC4-/- or TRPC(1-7)-/- mice, again revealing preferential interference with osteoclastogenesis driven by proinflammatory cytokines. ELP-004 also reduced bone erosion in a mouse model of rheumatoid arthritis. These investigations reveal TRPC channels as critical mediators of inflammatory bone erosion and provide insight into the major target of ELP-004, a drug currently in preclinical testing as a therapeutic for inflammatory arthritis.
Collapse
Affiliation(s)
- Suravi Ray
- Fels Cancer Institute for Personalized Medicine, Department of Cancer & Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jamie L. McCall
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- ExesaLibero Pharma, Morgantown, WV 26505, USA
| | - Jin Bin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston TX 77030, USA
| | - Jaepyo Jeon
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston TX 77030, USA
| | - Aidan Douglas
- Fels Cancer Institute for Personalized Medicine, Department of Cancer & Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Kendall Tyler
- Fels Cancer Institute for Personalized Medicine, Department of Cancer & Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Siyao Liu
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Kendyl Berry
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- ExesaLibero Pharma, Morgantown, WV 26505, USA
| | | | - Casey Hall
- ExesaLibero Pharma, Morgantown, WV 26505, USA
| | - Klaus Groschner
- Medical University of Graz, Division of Medical Physics and Biophysics, Neue Stiftingtalstrasse 6/H03, 8010 Graz, Austria
| | - Bernadett Bacsa
- Medical University of Graz, Division of Medical Physics and Biophysics, Neue Stiftingtalstrasse 6/H03, 8010 Graz, Austria
| | - Werner Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston TX 77030, USA
| | - Harry C. Blair
- Research Service, VA Medical Centre, Departments of Pathology and of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - John B. Barnett
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- ExesaLibero Pharma, Morgantown, WV 26505, USA
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
| | - Jonathan Soboloff
- Fels Cancer Institute for Personalized Medicine, Department of Cancer & Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
6
|
Iwanowska M, Kochman M, Szatko A, Zgliczyński W, Glinicki P. Bone Disease in Primary Hyperparathyroidism-Changes Occurring in Bone Metabolism and New Potential Treatment Strategies. Int J Mol Sci 2024; 25:11639. [PMID: 39519190 PMCID: PMC11546563 DOI: 10.3390/ijms252111639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Primary hyperparathyroidism (PHPT) is a common endocrinopathy, predominantly caused by a single parathyroid adenoma that is responsible for the excessive secretion of parathyroid hormone (PTH)-the hallmark of disease. Excess of this hormone causes remarkable changes in bone metabolism, including an increased level of bone remodeling with a predominance of bone resorption. Those changes lead to deterioration of bone structure and density, especially in cortical bone. The main treatment for PHPT is surgical removal of the adenoma, which normalizes PTH levels and terminates the progression of bone disease and leads to its regeneration. However, because not all the patients are suitable candidates for surgery, alternative therapies are needed. Current non-surgical treatments targeting bone disease secondary to PHPT include bisphosphonates and denosumab. Those antiresorptives prevent further bone loss, but they lack the ability to regenerate already degraded bone. There is ongoing research to find targeted drugs capable of halting resorption alongside stimulating bone formation. This review presents the advancements in understanding the molecular mechanisms responsible for bone disease in PHPT and assesses the efficacy of new potential therapeutic approaches (e.g., allosteric inhibitors of the PTH receptor, V-ATPase, or cathepsin inhibitors) aimed at mitigating bone loss and enhancing bone regeneration in affected patients.
Collapse
Affiliation(s)
- Mirella Iwanowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Magdalena Kochman
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Alicja Szatko
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- EndoLab Laboratory, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Wojciech Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Piotr Glinicki
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- EndoLab Laboratory, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| |
Collapse
|
7
|
Wu S, Xia Z, Wei L, Ji J, Zhang Y, Huang D. Secreted protein TNA: a promising biomarker for understanding the adipose-bone axis and its impact on bone metabolism. J Orthop Surg Res 2024; 19:610. [PMID: 39342371 PMCID: PMC11437659 DOI: 10.1186/s13018-024-05089-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Osteoporosis (OP) is a systemic bone disease characterized by reduced bone mass and deterioration of bone microstructure, leading to increased bone fragility. Platelets can take up and release cytokines, and a high platelet count has been associated with low bone density. Obesity is strongly associated with OP, and adipose tissue can influence platelet function by secreting adipokines. However, the biological relationship between these factors remains unclear. METHODS We conducted differential analysis to identify OP platelet-related plasma proteins. And, making comprehensive analysis, including functional enrichment, protein-protein interaction network analysis, and Friends analysis. The key protein, Tetranectin (TNA/CLEC3B), was identified through screening. Then, we analyzed TNA's potential roles in osteogenic and adipogenic differentiation using multiple RNA-seq data sets and validated its effect on osteoclast differentiation and bone resorption function through in vitro experiments. RESULTS Six OP-platelet-related proteins were identified via differential analysis. Then, we screened the key protein TNA, which was found to be highly expressed in adipose tissue. RNA-seq data suggested that TNA may promote early osteoblast differentiation. In vitro experiments showed that knockdown of TNA expression significantly increased the expression of osteoclast markers, thereby promoting osteoclast differentiation and bone resorption. CONCLUSIONS We identified TNA as a secreted protein that inhibits osteoclast differentiation and bone resorption. While, it potentially promoted early osteoblast differentiation from bioinformatic results. TNA may play a role in bone metabolism through the adipose-bone axis.
Collapse
Affiliation(s)
- Shaobo Wu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Zhihao Xia
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Liangliang Wei
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Jiajia Ji
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Dageng Huang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
8
|
Liu W, Yu W, Zhou L, Ling D, Xu Y, He F. Inhibition of ZDHHC16 promoted osteogenic differentiation and reduced ferroptosis of dental pulp stem cells by CREB. BMC Oral Health 2024; 24:388. [DOI: https:/doi.org/10.1186/s12903-024-04107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2025] Open
Abstract
Abstract
Background
The repair of bone defects caused by periodontal diseases is a difficult challenge in clinical treatment. Dental pulp stem cells (DPSCs) are widely studied for alveolar bone repair. The current investigation aimed to examine the specific mechanisms underlying the role of Zinc finger DHHC-type palmitoyl transferases 16 (ZDHHC16) in the process of osteogenic differentiation (OD) of DPSCs.
Methods
The lentiviral vectors ZDHHC16 or si-ZDHHC16 were introduced in the DPSCs and then the cells were induced by an odontogenic medium for 21 days. Subsequently, Quantitate Polymerase Chain Reaction (PCR), immunofluorescent staining, proliferation assay, ethynyl deoxyuridine (EdU) staining, and western blot analysis were used to investigate the specific details of ZDHHC16 contribution in OD of DPSCs.
Results
Our findings indicate that ZDHHC16 exhibited a suppressive effect on cellular proliferation and oxidative phosphorylation, while concurrently inducing ferroptosis in DPSCs. Moreover, the inhibition of ZDHHC16 promoted cell development and OD and reduced ferroptosis of DPSCs. The expression of p-CREB was suppressed by ZDHHC16, and immunoprecipitation (IP) analysis revealed that ZDHHC16 protein exhibited interconnection with cAMP-response element binding protein (CREB) of DPSCs. The CREB suppression reduced the impacts of ZDHHC16 on OD and ferroptosis of DPSCs. The activation of CREB also reduced the influences of si-ZDHHC16 on OD and ferroptosis of DPSCs.
Conclusions
These findings provide evidences to support a negative association between ZDHHC16 and OD of DPSCs, which might be mediated by ferroptosis of DPSCs via CREB.
Collapse
|
9
|
Liu W, Yu W, Zhou L, Ling D, Xu Y, He F. Inhibition of ZDHHC16 promoted osteogenic differentiation and reduced ferroptosis of dental pulp stem cells by CREB. BMC Oral Health 2024; 24:388. [PMID: 38532349 PMCID: PMC10964552 DOI: 10.1186/s12903-024-04107-x] [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: 07/09/2023] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND The repair of bone defects caused by periodontal diseases is a difficult challenge in clinical treatment. Dental pulp stem cells (DPSCs) are widely studied for alveolar bone repair. The current investigation aimed to examine the specific mechanisms underlying the role of Zinc finger DHHC-type palmitoyl transferases 16 (ZDHHC16) in the process of osteogenic differentiation (OD) of DPSCs. METHODS The lentiviral vectors ZDHHC16 or si-ZDHHC16 were introduced in the DPSCs and then the cells were induced by an odontogenic medium for 21 days. Subsequently, Quantitate Polymerase Chain Reaction (PCR), immunofluorescent staining, proliferation assay, ethynyl deoxyuridine (EdU) staining, and western blot analysis were used to investigate the specific details of ZDHHC16 contribution in OD of DPSCs. RESULTS Our findings indicate that ZDHHC16 exhibited a suppressive effect on cellular proliferation and oxidative phosphorylation, while concurrently inducing ferroptosis in DPSCs. Moreover, the inhibition of ZDHHC16 promoted cell development and OD and reduced ferroptosis of DPSCs. The expression of p-CREB was suppressed by ZDHHC16, and immunoprecipitation (IP) analysis revealed that ZDHHC16 protein exhibited interconnection with cAMP-response element binding protein (CREB) of DPSCs. The CREB suppression reduced the impacts of ZDHHC16 on OD and ferroptosis of DPSCs. The activation of CREB also reduced the influences of si-ZDHHC16 on OD and ferroptosis of DPSCs. CONCLUSIONS These findings provide evidences to support a negative association between ZDHHC16 and OD of DPSCs, which might be mediated by ferroptosis of DPSCs via CREB.
Collapse
Affiliation(s)
- Wei Liu
- Department of Oral Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
- Department of Oral Prosthodontics, Stomatology Hospital, School of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, 166 Qiu'tao Road (N), Hangzhou, Zhejiang, 310000, China
| | - Wenwei Yu
- Department of Oral Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Lili Zhou
- Department of Oral Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Danhua Ling
- Department of Oral Prosthodontics, Stomatology Hospital, School of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, 166 Qiu'tao Road (N), Hangzhou, Zhejiang, 310000, China
- Department of General Dentistry, the Second Affiliated Hospital of Zhejiang University School of Medicine, 1511 Jianghong Road, Hangzhou, Hangzhou, Zhejiang, 310052, China
| | - Yangbo Xu
- Department of Oral Prosthodontics, Stomatology Hospital, School of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, 166 Qiu'tao Road (N), Hangzhou, Zhejiang, 310000, China
| | - Fuming He
- Department of Oral Prosthodontics, Stomatology Hospital, School of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, 166 Qiu'tao Road (N), Hangzhou, Zhejiang, 310000, China.
| |
Collapse
|
10
|
Zheng L, Zhao S, Li Y, Xu J, Yan W, Guo B, Xu J, Jiang L, Zhang Y, Wei H, Jiang Q. Engineered MgO nanoparticles for cartilage-bone synergistic therapy. SCIENCE ADVANCES 2024; 10:eadk6084. [PMID: 38457498 PMCID: PMC10923500 DOI: 10.1126/sciadv.adk6084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
The emerging therapeutic strategies for osteoarthritis (OA) are shifting toward comprehensive approaches that target periarticular tissues, involving both cartilage and subchondral bone. This shift drives the development of single-component therapeutics capable of acting on multiple tissues and cells. Magnesium, an element essential for maintaining skeletal health, shows promise in treating OA. However, the precise effects of magnesium on cartilage and subchondral bone are not yet clear. Here, we investigated the therapeutic effect of Mg2+ on OA, unveiling its protective effects on both cartilage and bone at the cellular and animal levels. The beneficial effect on the cartilage-bone interaction is primarily mediated by the PI3K/AKT pathway. In addition, we developed poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with nano-magnesium oxide modified with stearic acid (SA), MgO&SA@PLGA, for intra-articular injection. These microspheres demonstrated remarkable efficacy in alleviating OA in rat models, highlighting their translational potential in clinical applications.
Collapse
Affiliation(s)
- Liming Zheng
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Sheng Zhao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Yixuan Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Wenjin Yan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Yifeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| |
Collapse
|
11
|
Yokota K. Osteoclast differentiation in rheumatoid arthritis. Immunol Med 2024; 47:6-11. [PMID: 37309864 DOI: 10.1080/25785826.2023.2220931] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
Osteoclasts, derived from the monocyte/macrophage line of bone marrow hematopoietic stem cell progenitors, are the sole bone-resorbing cells of the body. Conventional osteoclast differentiation requires macrophage colony-stimulating factor and receptor activator of nuclear factor kappa-B ligand (RANKL) signaling. Rheumatoid arthritis (RA) is the most prevalent systemic autoimmune disease and inflammatory arthritis characterized by bone destruction. Increased levels of proinflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), in the serum and joints, cause excessive bone destruction. We have recently reported that stimulation of human peripheral blood monocytes with TNF-α and IL-6 induces the differentiation of osteoclasts with bone resorption activity. This review presents the functional differences between representative osteoclasts, conventional RANKL-induced osteoclasts, and recently identified proinflammatory cytokine (TNF-α and IL-6)-induced osteoclasts in RA patients. We believe novel pathological osteoclasts associated with RA will be identified, and new therapeutic strategies will be developed to target these osteoclasts and prevent the progression of bone destruction.
Collapse
Affiliation(s)
- Kazuhiro Yokota
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
12
|
Huang Z, Peng Y, Ke G, Xiao Y, Chen Y. CaMKII may regulate renal tubular epithelial cell apoptosis through YAP/NFAT2 in acute kidney injury mice. Ren Fail 2023; 45:2172961. [PMID: 36718671 PMCID: PMC9891164 DOI: 10.1080/0886022x.2023.2172961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
AIM Renal tubular epithelial cell (RTEC) apoptosis is important in acute kidney injury (AKI). Calcium/calmodulin-dependent protein kinase II (CaMKII) plays an important role in cell apoptosis, but its potential role in AKI remains unknown. METHODS Using co-immunoprecipitation, immunofluorescence, immunohistochemistry, western blotting, flow cytometry, and cell transfection, this study aimed to verify whether CaMKII is involved in RTEC apoptosis and to explore the underlying mechanism. RESULTS We found that CaMKII was involved in RTEC apoptosis. In adriamycin-induced AKI mice, serum creatinine levels, cell apoptosis, CaMKII activity, and nuclear factor of activated T cells 2 (NFAT2) levels increased, whereas nuclear Yes-associated protein (YAP) expression decreased; inhibition of CaMKII activity reversed these changes. Phosphorylated CaMKII could bind to phosphorylated YAP in the cytoplasm and block it from entering the nucleus, thereby failing to inhibit NFAT2-mediated cell apoptosis. Sequestrated phosphorylated YAP in the RTEC cytoplasm was finally degraded by ubiquitination. CONCLUSION CaMKII may regulate RTEC apoptosis through YAP/NFAT2 in AKI mice. CaMKII may be a potent molecular target for AKI treatment.
Collapse
Affiliation(s)
- Zongshun Huang
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yonghua Peng
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guibao Ke
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun Xiao
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaqi Chen
- Department of Nephrology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
13
|
Luo Y, Liu H, Zhang Y, Liu Y, Liu S, Liu X, Luo E. Metal ions: the unfading stars of bone regeneration-from bone metabolism regulation to biomaterial applications. Biomater Sci 2023; 11:7268-7295. [PMID: 37800407 DOI: 10.1039/d3bm01146a] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
In recent years, bone regeneration has emerged as a remarkable field that offers promising guidance for treating bone-related diseases, such as bone defects, bone infections, and osteosarcoma. Among various bone regeneration approaches, the metal ion-based strategy has surfaced as a prospective candidate approach owing to the extensive regulatory role of metal ions in bone metabolism and the diversity of corresponding delivery strategies. Various metal ions can promote bone regeneration through three primary strategies: balancing the effects of osteoblasts and osteoclasts, regulating the immune microenvironment, and promoting bone angiogenesis. In the meantime, the complex molecular mechanisms behind these strategies are being consistently explored. Moreover, the accelerated development of biomaterials broadens the prospect of metal ions applied to bone regeneration. This review highlights the potential of metal ions for bone regeneration and their underlying mechanisms. We propose that future investigations focus on refining the clinical utilization of metal ions using both mechanistic inquiry and materials engineering to bolster the clinical effectiveness of metal ion-based approaches for bone regeneration.
Collapse
Affiliation(s)
- Yankun Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Emergency, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xian Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| |
Collapse
|
14
|
Omata Y, Tachibana H, Aizaki Y, Mimura T, Sato K. Essentiality of Nfatc1 short isoform in osteoclast differentiation and its self-regulation. Sci Rep 2023; 13:18797. [PMID: 37914750 PMCID: PMC10620225 DOI: 10.1038/s41598-023-45909-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
During osteoclast differentiation, the expression of the transcription factor nuclear factor of activated T cell 1 (Nfatc1) increases in an autoproliferative manner. Nfatc1 isoforms are of three sizes, and only the short isoform increases during osteoclast differentiation. Genetic ablation of the whole Nfatc1 gene demonstrated that it is essential for osteoclastogenesis; however, the specific role of the Nfatc1 short form (Nfatc1/αA) remains unknown. In this study, we engineered Nfatc1 short form-specific knockout mice and found that these mice died in utero by day 13.5. We developed a novel osteoclast culture system in which hematopoietic stem cells were cultured, proliferated, and then differentiated into osteoclasts in vitro. Using this system, we show that the Nfatc1/αA isoform is essential for osteoclastogenesis and is responsible for the expression of various osteoclast markers, the Nfatc1 short form itself, and Nfatc1 regulators.
Collapse
Affiliation(s)
- Yasuhiro Omata
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hideyuki Tachibana
- Department of Rheumatology, Akiru Municipal Medical Center, 78-1 Hikita, Akiruno, Tokyo, 197-0834, Japan
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Yoshimi Aizaki
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Toshihide Mimura
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Kojiro Sato
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
15
|
Jaberi S, Fahnestock M. Mechanisms of the Beneficial Effects of Exercise on Brain-Derived Neurotrophic Factor Expression in Alzheimer's Disease. Biomolecules 2023; 13:1577. [PMID: 38002258 PMCID: PMC10669442 DOI: 10.3390/biom13111577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a key molecule in promoting neurogenesis, dendritic and synaptic health, neuronal survival, plasticity, and excitability, all of which are disrupted in neurological and cognitive disorders such as Alzheimer's disease (AD). Extracellular aggregates of amyloid-β (Aβ) in the form of plaques and intracellular aggregates of hyperphosphorylated tau protein have been identified as major pathological insults in the AD brain, along with immune dysfunction, oxidative stress, and other toxic stressors. Although aggregated Aβ and tau lead to decreased brain BDNF expression, early losses in BDNF prior to plaque and tangle formation may be due to other insults such as oxidative stress and contribute to early synaptic dysfunction. Physical exercise, on the other hand, protects synaptic and neuronal structure and function, with increased BDNF as a major mediator of exercise-induced enhancements in cognitive function. Here, we review recent literature on the mechanisms behind exercise-induced BDNF upregulation and its effects on improving learning and memory and on Alzheimer's disease pathology. Exercise releases into the circulation a host of hormones and factors from a variety of peripheral tissues. Mechanisms of BDNF induction discussed here are osteocalcin, FNDC5/irisin, and lactate. The fundamental mechanisms of how exercise impacts BDNF and cognition are not yet fully understood but are a prerequisite to developing new biomarkers and therapies to delay or prevent cognitive decline.
Collapse
Affiliation(s)
- Sama Jaberi
- Graduate Program in Neuroscience, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
16
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
17
|
Pasupuleti SK, Ramdas B, Burns SS, Palam LR, Kanumuri R, Kumar R, Pandhiri TR, Dave UP, Yellapu NK, Zhou X, Zhang C, Sandusky GE, Yu Z, Honigberg MC, Bick AG, Griffin GK, Niroula A, Ebert BL, Paczesny S, Natarajan P, Kapur R. Obesity-induced inflammation exacerbates clonal hematopoiesis. J Clin Invest 2023; 133:e163968. [PMID: 37071471 PMCID: PMC10231999 DOI: 10.1172/jci163968] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
Characterized by the accumulation of somatic mutations in blood cell lineages, clonal hematopoiesis of indeterminate potential (CHIP) is frequent in aging and involves the expansion of mutated hematopoietic stem and progenitor cells (HSC/Ps) that leads to an increased risk of hematologic malignancy. However, the risk factors that contribute to CHIP-associated clonal hematopoiesis (CH) are poorly understood. Obesity induces a proinflammatory state and fatty bone marrow (FBM), which may influence CHIP-associated pathologies. We analyzed exome sequencing and clinical data for 47,466 individuals with validated CHIP in the UK Biobank. CHIP was present in 5.8% of the study population and was associated with a significant increase in the waist-to-hip ratio (WHR). Mouse models of obesity and CHIP driven by heterozygosity of Tet2, Dnmt3a, Asxl1, and Jak2 resulted in exacerbated expansion of mutant HSC/Ps due in part to excessive inflammation. Our results show that obesity is highly associated with CHIP and that a proinflammatory state could potentiate the progression of CHIP to more significant hematologic neoplasia. The calcium channel blockers nifedipine and SKF-96365, either alone or in combination with metformin, MCC950, or anakinra (IL-1 receptor antagonist), suppressed the growth of mutant CHIP cells and partially restored normal hematopoiesis. Targeting CHIP-mutant cells with these drugs could be a potential therapeutic approach to treat CH and its associated abnormalities in individuals with obesity.
Collapse
Affiliation(s)
| | - Baskar Ramdas
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and
| | - Sarah S. Burns
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and
| | | | - Rahul Kanumuri
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and
| | - Ramesh Kumar
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and
| | | | - Utpal P. Dave
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nanda Kumar Yellapu
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Xinyu Zhou
- Department of Medical and Molecular Genetics and
| | - Chi Zhang
- Department of Medical and Molecular Genetics and
| | - George E. Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhi Yu
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Michael C. Honigberg
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gabriel K. Griffin
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Epigenomics Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Abhishek Niroula
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Benjamin L. Ebert
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Medical University of South Carolina, Charlestown, South Carolina, USA
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Reuben Kapur
- Herman B Wells Center for Pediatric Research, Department of Pediatrics and
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
18
|
Fuiten AM, Yoshimoto Y, Shukunami C, Stadler HS. Digits in a dish: An in vitro system to assess the molecular genetics of hand/foot development at single-cell resolution. Front Cell Dev Biol 2023; 11:1135025. [PMID: 36994104 PMCID: PMC10040768 DOI: 10.3389/fcell.2023.1135025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
In vitro models allow for the study of developmental processes outside of the embryo. To gain access to the cells mediating digit and joint development, we identified a unique property of undifferentiated mesenchyme isolated from the distal early autopod to autonomously re-assemble forming multiple autopod structures including: digits, interdigital tissues, joints, muscles and tendons. Single-cell transcriptomic analysis of these developing structures revealed distinct cell clusters that express canonical markers of distal limb development including: Col2a1, Col10a1, and Sp7 (phalanx formation), Thbs2 and Col1a1 (perichondrium), Gdf5, Wnt5a, and Jun (joint interzone), Aldh1a2 and Msx1 (interdigital tissues), Myod1 (muscle progenitors), Prg4 (articular perichondrium/articular cartilage), and Scx and Tnmd (tenocytes/tendons). Analysis of the gene expression patterns for these signature genes indicates that developmental timing and tissue-specific localization were also recapitulated in a manner similar to the initiation and maturation of the developing murine autopod. Finally, the in vitro digit system also recapitulates congenital malformations associated with genetic mutations as in vitro cultures of Hoxa13 mutant mesenchyme produced defects present in Hoxa13 mutant autopods including digit fusions, reduced phalangeal segment numbers, and poor mesenchymal condensation. These findings demonstrate the robustness of the in vitro digit system to recapitulate digit and joint development. As an in vitro model of murine digit and joint development, this innovative system will provide access to the developing limb tissues facilitating studies to discern how digit and articular joint formation is initiated and how undifferentiated mesenchyme is patterned to establish individual digit morphologies. The in vitro digit system also provides a platform to rapidly evaluate treatments aimed at stimulating the repair or regeneration of mammalian digits impacted by congenital malformation, injury, or disease.
Collapse
Affiliation(s)
- Allison M. Fuiten
- Research Center, Shriners Children’s, Portland, OR, United States
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - H. Scott Stadler
- Research Center, Shriners Children’s, Portland, OR, United States
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
- *Correspondence: H. Scott Stadler,
| |
Collapse
|
19
|
Weng Y, Jian Y, Huang W, Xie Z, Zhou Y, Pei X. Alkaline earth metals for osteogenic scaffolds: From mechanisms to applications. J Biomed Mater Res B Appl Biomater 2023; 111:1447-1474. [PMID: 36883838 DOI: 10.1002/jbm.b.35246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023]
Abstract
Regeneration of bone defects is a significant challenge today. As alternative approaches to the autologous bone, scaffold materials have remarkable features in treating bone defects; however, the various properties of current scaffold materials still fall short of expectations. Due to the osteogenic capability of alkaline earth metals, their application in scaffold materials has become an effective approach to improving their properties. Furthermore, numerous studies have shown that combining alkaline earth metals leads to better osteogenic properties than applying them alone. In this review, the physicochemical and physiological characteristics of alkaline earth metals are introduced, mainly focusing on their mechanisms and applications in osteogenesis, especially magnesium (Mg), calcium (Ca), strontium (Sr), and barium (Ba). Furthermore, this review highlights the possible cross-talk between pathways when alkaline earth metals are combined. Finally, some of the current drawbacks of scaffold materials are enumerated, such as the high corrosion rate of Mg scaffolds and defects in the mechanical properties of Ca scaffolds. Moreover, a brief perspective is also provided regarding future directions in this field. It is worth exploring that whether the levels of alkaline earth metals in newly regenerated bone differs from those in normal bone. The ideal ratio of each element in the bone tissue engineering scaffolds or the optimal concentration of each elemental ion in the created osteogenic environment still needs further exploration. The review not only summarizes the research developments in osteogenesis but also offers a direction for developing new scaffold materials.
Collapse
Affiliation(s)
- Yihang Weng
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Yujia Jian
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Wenlong Huang
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhuojun Xie
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Zhou
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Xibo Pei
- Department of Prosthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
20
|
Nam MH, Park HJ, Seo YK. Reduction of Osteoclastic Differentiation of Raw 264.7 Cells by EMF Exposure through TRPV4 and p-CREB Pathway. Int J Mol Sci 2023; 24:ijms24043058. [PMID: 36834470 PMCID: PMC9959640 DOI: 10.3390/ijms24043058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/28/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
In this study, we investigated the effect of EMF exposure on the regulation of RANKL-induced osteoclast differentiation in Raw 264.7 cells. In the EMF-exposed group, the cell volume did not increase despite RANKL treatment, and the expression levels of Caspase-3 remained much lower than those in the RANKL-treated group. TRAP and F-actin staining revealed smaller actin rings in cells exposed to EMF during RANKL-induced differentiation, indicating that EMF inhibited osteoclast differentiation. EMF-irradiated cells exhibited reduced mRNA levels of osteoclastic differentiation markers cathepsin K (CTSK), tartrate-resistant acid phosphatase (TRAP), and matrix metalloproteinase 9 (MMP-9). Furthermore, as measured by RT-qPCR and Western blot, EMF induced no changes in the levels of p-ERK and p-38; however, it reduced the levels of TRPV4 and p-CREB. Overall, our findings indicate that EMF irradiation inhibits osteoclast differentiation through the TRPV4 and p-CREB pathway.
Collapse
|
21
|
Koga T, Umeda M, Yoshida N, Satyam A, Jha M, Scherlinger M, Bhargava R, Tsokos MG, Sato T, Furukawa K, Endo Y, Fukui S, Iwamoto N, Abiru N, Okita M, Ito M, Kawakami A, Tsokos GC. Inhibition of calcium/calmodulin-dependent protein kinase IV in arthritis: dual effect on Th17 cell activation and osteoclastogenesis. Rheumatology (Oxford) 2023; 62:861-871. [PMID: 35781320 PMCID: PMC9891404 DOI: 10.1093/rheumatology/keac381] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 05/06/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To investigate the role of calcium/calmodulin-dependent protein kinase IV (CaMK4) in the development of joint injury in a mouse model of arthritis and patients with RA. METHODS Camk4-deficient, Camk4flox/floxLck-Cre, and mice treated with CaMK4 inhibitor KN-93 or KN-93 encapsulated in nanoparticles tagged with CD4 or CD8 antibodies were subjected to collagen-induced arthritis (CIA). Inflammatory cytokine levels, humoral immune response, synovitis, and T-cell activation were recorded. CAMK4 gene expression was measured in CD4+ T cells from healthy participants and patients with active RA. Micro-CT and histology were used to assess joint pathology. CD4+ and CD14+ cells in patients with RA were subjected to Th17 or osteoclast differentiation, respectively. RESULTS CaMK4-deficient mice subjected to CIA displayed improved clinical scores and decreased numbers of Th17 cells. KN-93 treatment significantly reduced joint destruction by decreasing the production of inflammatory cytokines. Furthermore, Camk4flox/floxLck-Cre mice and mice treated with KN93-loaded CD4 antibody-tagged nanoparticles developed fewer Th17 cells and less severe arthritis. CaMK4 inhibition mitigated IL-17 production by CD4+ cells in patients with RA. The number of in vitro differentiated osteoclasts from CD14+ cells in patients with RA was significantly decreased with CaMK4 inhibitors. CONCLUSION Using global and CD4-cell-targeted pharmacologic approaches and conditionally deficient mice, we demonstrate that CaMK4 is important in the development of arthritis. Using ex vivo cell cultures from patients with RA, CaMK4 is important for both Th17 generation and osteoclastogenesis. We propose that CaMK4 inhibition represents a new approach to control the development of arthritis.
Collapse
Affiliation(s)
- Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences.,Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Masataka Umeda
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences.,Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nobuya Yoshida
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Abhigyan Satyam
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Meenakshi Jha
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marc Scherlinger
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Rhea Bhargava
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria G Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Tomohito Sato
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences
| | - Kaori Furukawa
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences
| | - Yushiro Endo
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences
| | - Shoichi Fukui
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences
| | - Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences
| | - Norio Abiru
- Department of Endocrinology and Metabolism, Division of Advanced Preventive Medical Sciences
| | - Minoru Okita
- Department of Physical Therapy Science, Unit of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | - Masako Ito
- Nagasaki Study Center, The Open University of Japan, Chiba, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Yu BF, Wang Z, Chen XX, Zeng Q, Dai CC, Wei J. Continuous dynamic identification of key genes and molecular signaling pathways of periosteum in guided bone self-generation in swine model. J Orthop Surg Res 2023; 18:53. [PMID: 36653843 PMCID: PMC9847205 DOI: 10.1186/s13018-023-03524-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Guided bone self-generation with periosteum-preserved has successfully regenerated mandibular, temporomandibular and interphalangeal joint. The aim of this study was to investigate the dynamic changes of gene expression of periosteum which was involved in the guided bone self-generation. METHODS Rib defects of critical size were created in mature swine with periosteum-preserved. The periosteum was sutured into a sealed sheath that closed the bone defect. The periosteum of trauma and control sites were harvested at postoperative 9 time points, and total RNA was extracted. Microarray analysis was conducted to identify the differences in the transcriptome of different time points between two groups. RESULTS The differentially expressed genes (DEGs) between control and trauma group were different at postoperative different time points. The dynamic changes of the number of DEGs fluctuated a lot. There were 3 volatility peaks, and we chose 3 time points of DEG number peak (1 week, 5 weeks and 6 months) to study the functions of DEGs. Oxidoreductase activity, oxidation-reduction process and mitochondrion are the most enriched terms of Go analysis. The major signaling pathways of DEGs enrichment include oxidative phosphorylation, PI3K-Akt signaling pathway, osteoclast differentiation pathway and Wnt signaling. CONCLUSIONS The oxidoreductase reaction was activated during this bone regeneration process. The oxidative phosphorylation, PI3K-Akt signaling pathway, osteoclast differentiation pathway and Wnt signaling may play important roles in the guided bone self-generation with periosteum-preserved. This study can provide a reference for how to improve the application of this concept of bone regeneration.
Collapse
Affiliation(s)
- Bao-Fu Yu
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Zi Wang
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Xiao-Xue Chen
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Qi Zeng
- grid.415002.20000 0004 1757 8108Department of Plastic Surgery, Jiangxi Province People’s Hospital, Nanchang, China
| | - Chuan-Chang Dai
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| | - Jiao Wei
- grid.412523.30000 0004 0386 9086Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011 China
| |
Collapse
|
23
|
Hansen MS, Søe K, Christensen LL, Fernandez-Guerra P, Hansen NW, Wyatt RA, Martin C, Hardy RS, Andersen TL, Olesen JB, Hartmann B, Rosenkilde MM, Kassem M, Rauch A, Gorvin CM, Frost M. GIP reduces osteoclast activity and improves osteoblast survival in primary human bone cells. Eur J Endocrinol 2023; 188:6987865. [PMID: 36747334 DOI: 10.1093/ejendo/lvac004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/26/2022] [Accepted: 11/19/2022] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Drugs targeting the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) are emerging as treatments for type-2 diabetes and obesity. GIP acutely decreases serum markers of bone resorption and transiently increases bone formation markers in short-term clinical investigations. However, it is unknown whether GIP acts directly on bone cells to mediate these effects. Using a GIPR-specific antagonist, we aimed to assess whether GIP acts directly on primary human osteoclasts and osteoblasts. METHODS Osteoclasts were differentiated from human CD14+ monocytes and osteoblasts from human bone. GIPR expression was determined using RNA-seq in primary human osteoclasts and in situ hybridization in human femoral bone. Osteoclastic resorptive activity was assessed using microscopy. GIPR signaling pathways in osteoclasts and osteoblasts were assessed using LANCE cAMP and AlphaLISA phosphorylation assays, intracellular calcium imaging and confocal microscopy. The bioenergetic profile of osteoclasts was evaluated using Seahorse XF-96. RESULTS GIPR is robustly expressed in mature human osteoclasts. GIP inhibits osteoclastogenesis, delays bone resorption, and increases osteoclast apoptosis by acting upon multiple signaling pathways (Src, cAMP, Akt, p38, Akt, NFκB) to impair nuclear translocation of nuclear factor of activated T cells-1 (NFATc1) and nuclear factor-κB (NFκB). Osteoblasts also expressed GIPR, and GIP improved osteoblast survival. Decreased bone resorption and improved osteoblast survival were also observed after GIP treatment of osteoclast-osteoblast co-cultures. Antagonizing GIPR with GIP(3-30)NH2 abolished the effects of GIP on osteoclasts and osteoblasts. CONCLUSIONS GIP inhibits bone resorption and improves survival of human osteoblasts, indicating that drugs targeting GIPR may impair bone resorption, whilst preserving bone formation.
Collapse
Affiliation(s)
- Morten S Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham B15 2TT, United Kingdom
| | - Kent Søe
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense C DK-5000, Denmark
| | - Line L Christensen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Paula Fernandez-Guerra
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Nina W Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Rachael A Wyatt
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham B15 2TT, United Kingdom
| | - Claire Martin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Rowan S Hardy
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Thomas L Andersen
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense C DK-5000, Denmark
| | - Jacob B Olesen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
| | - Alexander Rauch
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Steno Diabetes Centre Odense, Odense University Hospital, Odense C DK-5000, Denmark
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham B15 2TT, United Kingdom
| | - Morten Frost
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Steno Diabetes Centre Odense, Odense University Hospital, Odense C DK-5000, Denmark
| |
Collapse
|
24
|
ZDHHC16 restrains osteogenic differentiation of bone marrow mesenchymal stem cells by inhibiting phosphorylation of CREB. Heliyon 2023; 9:e12788. [PMID: 36685387 PMCID: PMC9852670 DOI: 10.1016/j.heliyon.2022.e12788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
Aims The osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs) plays a critical role in fracture healing. Osteogenic differentiation is regulated by a variety of post-translational modifications, but the function of protein palmitoylation in osteogenesis remains largely unknown. Methods Osteogenic differentiation induction of hBMSCs was used in this study. RT‒qPCR and immunoblotting assays (WB) were used to test marker genes of osteogenic induction. Alkaline phosphatase (ALP) activity, ALP staining and Alizarin red staining were performed to evaluate osteogenesis of hBMSCs. Signal finder pathway reporter array, co-immunoprecipitation and WB were applied to elucidate the molecular mechanism. A mouse fracture model was used to verify the in vivo function of the ZDHHC inhibitor. Key findings We revealed that palmitic acid inhibited Runx2 mRNA expression in hBMSCs and identified ZDHHC16 as a potential target palmitoyl acyltransferase. In addition, ZDHHC16 decreased during osteogenic induction. Next, we confirmed the inhibitory function of ZDHHC16 by its knockdown or overexpression during osteogenesis of hBMSCs. Moreover, we illustrated that ZDHHC16 inhibited the phosphorylation of CREB, thus inhibiting osteogenesis of hBMSCs by enhancing the palmitoylation of CREB. With a mouse femur fracture model, we found that 2-BP, a general inhibitor of ZDHHCs, promoted fracture healing in vivo. Thus, we clarified the inhibitory function of ZDHHC16 during osteogenic differentiation. Significance Collectively, these findings highlight the inhibitory function of ZDHHC16 in osteogenesis as a potential therapy method for fracture healing.
Collapse
|
25
|
Li Q, Wang R, Zhang Z, Wang H, Lu X, Zhang J, Kong APS, Tian XY, Chan HF, Chung ACK, Cheng JCY, Jiang Q, Lee WYW. Sirt3 mediates the benefits of exercise on bone in aged mice. Cell Death Differ 2023; 30:152-167. [PMID: 36153410 PMCID: PMC9883264 DOI: 10.1038/s41418-022-01053-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023] Open
Abstract
Exercise in later life is important for bone health and delays the progression of osteoporotic bone loss. Osteocytes are the major bone cells responsible for transforming mechanical stimuli into cellular signals through their highly specialized lacunocanalicular networks (LCN). Osteocyte activity and LCN degenerate with aging, thus might impair the effectiveness of exercise on bone health; however, the underlying mechanism and clinical implications remain elusive. Herein, we showed that deletion of Sirt3 in osteocytes could impair the formation of osteocyte dendritic processes and inhibit bone gain in response to exercise in vivo. Mechanistic studies revealed that Sirt3 regulates E11/gp38 through the protein kinase A (PKA)/cAMP response element-binding protein (CREB) signaling pathway. Additionally, the Sirt3 activator honokiol enhanced the sensitivity of osteocytes to fluid shear stress in vitro, and intraperitoneal injection of honokiol reduced bone loss in aged mice in a dose-dependent manner. Collectively, Sirt3 in osteocytes regulates bone mass and mechanical responses through the regulation of E11/gp38. Therefore, targeting Sirt3 could be a novel therapeutic strategy to prevent age-related bone loss and augment the benefits of exercise on the senescent skeleton.
Collapse
Affiliation(s)
- Qiangqiang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Rongliang Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhe Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaomin Lu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jiajun Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Alice Pik-Shan Kong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hon-Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Arthur Chi-Kong Chung
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jack Chun-Yiu Cheng
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
- SH Ho Scoliosis Research Laboratory, Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing University, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
26
|
Electroactive Hydroxyapatite/Carbon Nanofiber Scaffolds for Osteogenic Differentiation of Human Adipose-Derived Stem Cells. Int J Mol Sci 2022; 24:ijms24010530. [PMID: 36613973 PMCID: PMC9820130 DOI: 10.3390/ijms24010530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Traditional bone defect treatments are limited by an insufficient supply of autologous bone, the immune rejection of allogeneic bone grafts, and high medical costs. To address this medical need, bone tissue engineering has emerged as a promising option. Among the existing tissue engineering materials, the use of electroactive scaffolds has become a common strategy in bone repair. However, single-function electroactive scaffolds are not sufficient for scientific research or clinical application. On the other hand, multifunctional electroactive scaffolds are often complicated and expensive to prepare. Therefore, we propose a new tissue engineering strategy that optimizes the electrical properties and biocompatibility of carbon-based materials. Here, a hydroxyapatite/carbon nanofiber (HAp/CNF) scaffold with optimal electrical activity was prepared by electrospinning HAp nanoparticle-incorporated polyvinylidene fluoride (PVDF) and then carbonizing the fibers. Biochemical assessments of the markers of osteogenesis in human adipose-derived stem cells (h-ADSCs) cultured on HAp/CNF scaffolds demonstrate that the material promoted the osteogenic differentiation of h-ADSCs in the absence of an osteogenic factor. The results of this study show that electroactive carbon materials with a fibrous structure can promote the osteogenic differentiation of h-ADSCs, providing a new strategy for the preparation and application of carbon-based materials in bone tissue engineering.
Collapse
|
27
|
Niu Q, Gao J, Wang L, Liu J, Zhang L. Regulation of differentiation and generation of osteoclasts in rheumatoid arthritis. Front Immunol 2022; 13:1034050. [PMID: 36466887 PMCID: PMC9716075 DOI: 10.3389/fimmu.2022.1034050] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 09/25/2023] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA), which affects nearly 1% of the world's population, is a debilitating autoimmune disease. Bone erosion caused by periarticular osteopenia and synovial pannus formation is the most destructive pathological changes of RA, also leads to joint deformity and loss of function,and ultimately affects the quality of life of patients. Osteoclasts (OCs) are the only known bone resorption cells and their abnormal differentiation and production play an important role in the occurrence and development of RA bone destruction; this remains the main culprit behind RA. METHOD Based on the latest published literature and research progress at home and abroad, this paper reviews the abnormal regulation mechanism of OC generation and differentiation in RA and the possible targeted therapy. RESULT OC-mediated bone destruction is achieved through the regulation of a variety of cytokines and cell-to-cell interactions, including gene transcription, epigenetics and environmental factors. At present, most methods for the treatment of RA are based on the regulation of inflammation, the inhibition of bone injury and joint deformities remains unexplored. DISCUSSION This article will review the mechanism of abnormal differentiation of OC in RA, and summarise the current treatment oftargeting cytokines in the process of OC generation and differentiation to reduce bone destruction in patients with RA, which isexpected to become a valuable treatment choice to inhibit bone destruction in RA.
Collapse
Affiliation(s)
- Qing Niu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiaxi Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
28
|
Chanpaisaeng K, Reyes‐Fernandez PC, Dilkes B, Fleet JC. Diet X Gene Interactions Control Femoral Bone Adaptation to Low Dietary Calcium. JBMR Plus 2022; 6:e10668. [PMID: 36111202 PMCID: PMC9465001 DOI: 10.1002/jbm4.10668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/29/2022] [Accepted: 07/22/2022] [Indexed: 11/12/2022] Open
Abstract
Genetics and dietary calcium (Ca) are each critical regulators of peak bone mass but it is unclear how genetics alters the physiologic response of bone to dietary Ca restriction (RCR). Here, we conducted genetic mapping in C57BL/6J × DBA/2J (BXD) recombinant inbred mouse lines to identify environmentally sensitive loci controlling whole-bone mass (bone mineral density [BMD], bone mineral content [BMC]), distal trabecular bone, and cortical bone midshaft of the femur. Mice were fed adequate (basal) or low Ca diets from 4-12 weeks of age. Femurs were then examined by dual-energy X-ray absorptiometry (DXA) and micro-computed tomography (μCT). Body size-corrected residuals were used for statistical analysis, genetic mapping, and to estimate narrow sense heritability (h2). Genetics had a strong impact on femoral traits (eg, bone volume fraction [BV/TV] basal Ca, h2 = 0.60) as well as their RCR (eg, BV/TV, h2 = 0.32). Quantitative trait locus (QTL) mapping identified up to six loci affecting each bone trait. A subset of loci was detected in both diet groups, providing replication of environmentally robust genetic effects. Several loci control multiple bone phenotypes suggesting the existence of genetic pleiotropy. QTL controlling the bone RCR did not overlap with basal diet QTL, demonstrating genetic independence of those traits. Candidate genes underlying select multi-trait loci were prioritized by protein coding effects or gene expression differences in bone cells. These include candidate alleles in Rictor (chromosome [chr] 15) and Egfl7 (chr 2) at loci affecting bone in the basal or low Ca groups and in Msr1 (chr 8), Apc, and Camk4 (chr 18) at loci affecting RCR. By carefully controlling dietary Ca and measuring traits in age-matched mice we identified novel genetic loci determining bone mass/microarchitecture of the distal femur as well as their physiologic adaptation to inadequate dietary Ca intake. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Krittikan Chanpaisaeng
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA)Pathum ThaniThailand
| | - Perla C. Reyes‐Fernandez
- School of Health and Human Sciences, Department of Physical TherapyIndiana University–Purdue University IndianapolisIndianapolisINUSA
| | - Brian Dilkes
- Center for Plant BiologyPurdue UniversityWest LafayetteINUSA
- Department of BiochemistryPurdue UniversityWest LafayetteINUSA
| | - James C. Fleet
- Department of Nutritional Sciences and the Dell Pediatric Research InstituteUniversity of TexasAustinTXUSA
| |
Collapse
|
29
|
Zhang J, Zhang D, Zhao J. CFNAs of RBCs affect the release of inflammatory factors through the expression of CaMKIV in macrophages. Transfus Apher Sci 2022; 61:103494. [PMID: 35773126 DOI: 10.1016/j.transci.2022.103494] [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: 04/07/2022] [Revised: 06/11/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Blood transfusions reportedly modulate the recipient's immune system. Transfusion-related immunomodulation has been suggested as a mechanism of some adverse clinical outcomes. Extracellular nucleic acids circulate in plasma and activate relevant immune responses, but little is known about their mechanism of action in transfusion-related immunomodulation (TRIM). The aim of this study was to investigate the effects of cell-free nucleic acids (CFNAs) produced by red blood cells (RBCs) on innate immunity, especially peripheral blood mononuclear cells (PBMCs) and macrophages, and to investigate the mechanism of action. METHODS Differentially expressed genes (DEGs) between PBMCs exposed to RBC-produced CFNA and normal PBMCs were analyzed by gene expression data combined with bioinformatics. KEGG and GO enrichment analyses were performed for the DEGs, and in vitro experiments were performed for the effects of key genes on the release of inflammatory factors from macrophages. RESULTS Analysis of microarray data showed that exposure of monocytes to RBC-produced CFNAs increased the expression of genes involved in the innate immune response, including chemokines, chemokine receptors, and innate response receptors, and that calcium channel activity was highly regulated, with a key gene being CaMKIV. CaMKIV played a critical role in LPS-induced inflammatory factor release from macrophages, which was exacerbated by overexpression of the CaMKIV gene. CONCLUSION RBCs regulate the release of inflammatory factors during blood transfusion by releasing CFNAs and affecting expression of the CaMKIV gene in PBMCs or macrophages, which is a potential regulatory mechanism of blood transfusion-related immune regulation and related adverse reactions.
Collapse
Affiliation(s)
- Jingrui Zhang
- Department of Transfusion Medicine, General Hospital of Northern Theater Command, Shenyang 110000, China.
| | - Dan Zhang
- Department of Transfusion Medicine, General Hospital of Northern Theater Command, Shenyang 110000, China
| | - Jing Zhao
- Department of Transfusion Medicine, General Hospital of Northern Theater Command, Shenyang 110000, China
| |
Collapse
|
30
|
Dong C, Liu X, Li J, Lan D, Zheng S. Dysregulation of the HOTAIR-miR-152-CAMKIIα Axis in Craniosynostosis Results in Impaired Osteoclast Differentiation. Front Genet 2022; 13:787734. [PMID: 35360844 PMCID: PMC8961285 DOI: 10.3389/fgene.2022.787734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/21/2022] [Indexed: 01/17/2023] Open
Abstract
Craniosynostosis is one of the most common craniofacial deformities demanding surgical treatment in infancy. LncRNA HOTAIR has verified its important role in osteogenesis and osteoarthritis. However, whether HOTAIR plays an essential role in the development of craniosynostosis is still unclear. In this study, we aimed to investigate the molecular role of HOTAIR in the osteoclast function and development of craniosynostosis.For osteoclast differentiation, RAW264.7 cells were induced by 50 ng/ml of RANKL and 10 ng/mL M-CSF, followed by TRAP staining. Cell proliferation and apoptosis were assayed by the CCK-8 kit and Annexin V-FITC apoptosis detection kit, respectively. The expression of HOTAIR was determined in PBMCs by qRT-PCR. Protein levels of all those involved genes were measured by Western blot assay. A luciferase reporter assay was used to determine the miRNA target validation. The HOTAIR expression in PBMCs from children with craniosynostosis was significantly downregulated. The results of cell proliferation and apoptosis assays indicated that silencing of HOTAIR could inhibit osteoclast differentiation and increase cell apoptosis. Moreover, the luciferase reporter assay revealed that the regulatory axis and HOTAIR-miR-152-CAMKIIα were the regulatory mechanisms of HOTAIR in the osteoclast function and development of craniosynostosis.In this study, our data showed that HOTAIR could promote osteoclast differentiation by binding miR-152. Furthermore, the HOTAIR/HOTAIR-miR-152-CAMKIIα axis was found to regulate osteoclast differentiation. These results indicate that the HOTAIR plays a crucial role in the development of osteoclasts.
Collapse
Affiliation(s)
| | | | | | | | - Shan Zheng
- Department of Plastic Surgery, Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
31
|
Tang W, Wang H, Zhao X, Liu S, Kong SK, Ho A, Chen T, Feng H, He H. Stem cell differentiation with consistent lineage commitment induced by a flash of ultrafast-laser activation in vitro and in vivo. Cell Rep 2022; 38:110486. [PMID: 35263591 DOI: 10.1016/j.celrep.2022.110486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 12/06/2021] [Accepted: 02/14/2022] [Indexed: 11/03/2022] Open
Abstract
Recent technological advancements on stem cell differentiation induction have been making great progress in stem cell research, regenerative medicine, and therapeutic applications. However, the risk of off-target differentiation limits the wide application of stem cell therapy strategies. Here, we report a non-invasive all-optical strategy to induce stem cell differentiation in vitro and in vivo that activates individual target stem cells in situ by delivering a transient 100-ms irradiation of a tightly focused femtosecond laser to a submicron cytoplasmic region of primary adipose-derived stem cells (ADSCs). The ADSCs differentiate to osteoblasts with stable lineage commitment that cannot further transdifferentiate because of simultaneous initiation of multiple signaling pathways through specific Ca2+ kinetic patterns. This method can work in vivo to direct mouse cerebellar granule neuron progenitors to granule neurons in intact mouse cerebellums through the skull. Hence, this optical method without any genetic manipulations or exogenous biomaterials holds promising potential in biomedical research and cell-based therapies.
Collapse
Affiliation(s)
- Wanyi Tang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Haipeng Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiaohui Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Shiyue Liu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, P.R. China
| | - Siu Kai Kong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, P.R. China
| | - Aaron Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P.R. China
| | - Tunan Chen
- Institute of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Hua Feng
- Institute of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P.R. China
| | - Hao He
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P.R. China.
| |
Collapse
|
32
|
Hu W, Yu Y, Sun Y, Yuan F, Zhao F. MiR-25 overexpression inhibits titanium particle-induced osteoclast differentiation via down-regulation of mitochondrial calcium uniporter in vitro. J Orthop Surg Res 2022; 17:133. [PMID: 35241114 PMCID: PMC8895597 DOI: 10.1186/s13018-022-03030-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 02/18/2022] [Indexed: 11/10/2022] Open
Abstract
Background Mitochondrial calcium uniporter (MCU) is an important ion channel regulating calcium transport across the mitochondrial membrane. Calcium signaling, particularly via the Ca2+/NFATc1 pathway, has been identified as an important mediator of the osteoclast differentiation that leads to osteolysis around implants. The present study aimed to investigate whether down-regulation of MCU using microRNA-25 (miR-25) mimics could reduce osteoclast differentiation induced upon exposure to titanium (Ti) particles. Methods Ti particles were prepared. Osteoclast differentiation of RAW264.7 cells was induced by adding Ti particles and determined by TRAP staining. Calcium oscillation was determined using a dual-wavelength technique. After exposure of the cells in each group to Ti particles or control medium for 5 days, relative MCU and NFATc1 mRNA expression levels were determined by RT-qPCR. MCU and NFATc1 protein expression was determined by western blotting. NFATc1 activation was determined by immunofluorescence staining. Comparisons among multiple groups were conducted using one-way analysis of variance followed by Tukey test, and differences were considered significant if p < 0.05. Results MCU expression was reduced in response to miR-25 overexpression during the process of RAW 264.7 cell differentiation induced by Ti particles. Furthermore, osteoclast formation was inhibited, as evidenced by the low amplitude of calcium ion oscillation, reduced NFATc1 activation, and decreased mRNA and protein expression levels of nuclear factor-κB p65 and calmodulin kinases II/IV. Conclusions Regulation of MCU expression can impact osteoclast differentiation, and the underlying mechanism likely involves the Ca2+/NFATc1 signal pathway. Therefore, MCU may be a promising target in the development of new strategies to prevent and treat periprosthetic osteolysis. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-022-03030-7.
Collapse
Affiliation(s)
- Weifan Hu
- Department of Orthopedics, The People's Hospital of Jiawang District of Xuzhou, Xuzhou, 221000, People's Republic of China.,Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Quanshan District, Xuzhou City, Jiangsu Province, 221000, People's Republic of China
| | - Yongbo Yu
- Department of Orthopedics, The People's Hospital of Jiawang District of Xuzhou, Xuzhou, 221000, People's Republic of China
| | - Yang Sun
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Quanshan District, Xuzhou City, Jiangsu Province, 221000, People's Republic of China
| | - Feng Yuan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Quanshan District, Xuzhou City, Jiangsu Province, 221000, People's Republic of China
| | - Fengchao Zhao
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Quanshan District, Xuzhou City, Jiangsu Province, 221000, People's Republic of China.
| |
Collapse
|
33
|
Wang G, Ma C, Chen K, Wang Z, Qiu H, Chen D, He J, Zhang C, Guo D, Lai B, Zhang S, Huang L, Yang F, Yuan J, Chen L, He W, Xu J. Cycloastragenol Attenuates Osteoclastogenesis and Bone Loss by Targeting RANKL-Induced Nrf2/Keap1/ARE, NF-κB, Calcium, and NFATc1 Pathways. Front Pharmacol 2022; 12:810322. [PMID: 35126144 PMCID: PMC8812338 DOI: 10.3389/fphar.2021.810322] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/20/2021] [Indexed: 10/21/2023] Open
Abstract
Osteoporosis, which typically affects postmenopausal women, is an osteolytic disease due to over-activation of osteoclasts. However, current drugs targeting osteoclast inhibition face various side effects, making natural compounds with great interest as alternative treatment options. Cycloastragenol (CAG) is a triterpenoid with multiple biological activities. Previously, CAG's activity against aging-related osteoporosis was reported, but the mechanisms of actions for the activities were not understood. This study demonstrated that CAG dose-dependently inhibited osteoclast formation in receptor activator of nuclear factor-κB ligand (RANKL)-stimulated bone marrow macrophage (BMMs). Mechanism studies showed that CAG inhibited NF-κB, calcium, and nuclear factor of activated T cells 1 (NFATc1) pathways. Additionally, CAG also promoted the nuclear factor-erythroid 2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1)/anti-oxidative response element (ARE) pathway that scavenges reactive oxygen species (ROS). Furthermore, CAG was also found to prevent bone loss of postmenopausal osteoporosis (PMO) in a preclinical model of ovariectomized (OVX) mice. Collectively, our research confirms that CAG inhibits the formation and function of osteoclasts by regulating RANKL-induced intracellular signaling pathways, which may represent a promising alternative for the therapy of osteoclast-related disease.
Collapse
Affiliation(s)
- Gang Wang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Chao Ma
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Kai Chen
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Ziyi Wang
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Heng Qiu
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Delong Chen
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jianbo He
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Cheng Zhang
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Ding Guo
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Boyong Lai
- Guangzhou University of Chinese Medicine, Guangdong, China
| | | | - Linfeng Huang
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Fan Yang
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jinbo Yuan
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Leilei Chen
- Guangzhou University of Chinese Medicine, Guangdong, China
- Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Wei He
- Guangzhou University of Chinese Medicine, Guangdong, China
- Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| |
Collapse
|
34
|
Wang P, Zhang Z, Yin B, Li J, Xialin C, Lian W, Su Y, Jia C. Identifying changes in immune cells and constructing prognostic models using immune-related genes in post-burn immunosuppression. PeerJ 2022; 10:e12680. [PMID: 35070500 PMCID: PMC8761370 DOI: 10.7717/peerj.12680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/02/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Burn patients are prone to infection as well as immunosuppression, which is a significant cause of death. Currently, there is a lack of prognostic biomarkers for immunosuppression in burn patients. This study was conducted to identify immune-related genes that are prognosis biomarkers in post-burn immunosuppression and potential targets for immunotherapy. METHODS We downloaded the gene expression profiles and clinical data of 213 burn patients and 79 healthy samples from the Gene Expression Omnibus (GEO) database. Immune infiltration analysis was used to identify the proportion of circulating immune cells. Functional enrichment analyses were carried out to identify immune-related genes that were used to build miRNA-mRNA networks to screen key genes. Next, we carried out correlation analysis between immune cells and key genes that were then used to construct logistic regression models in GSE77791 and were validated in GSE19743. Finally, we determined the expression of key genes in burn patients using quantitative reverse transcription polymerase chain reaction (qRT-PCR). RESULTS A total of 745 differently expressed genes were screened out: 299 were up-regulated and 446 were down-regulated. The number of Th-cells (CD4+) decreased while neutrophils increased in burn patients. The enrichment analysis showed that down-regulated genes were enriched in the T-cell activation pathway, while up-regulated genes were enriched in neutrophil activation response in burn patients. We screened out key genes (NFATC2, RORA, and CAMK4) that could be regulated by miRNA. The expression of key genes was related to the proportion of Th-cells (CD4+) and survival, and was an excellent predictor of prognosis in burns with an area under the curve (AUC) value of 0.945. Finally, we determined that NFATC2, RORA, and CAMK4 were down-regulated in burn patients. CONCLUSION We found that NFATC2, RORA, and CAMK4 were likely prognostic biomarkers in post-burn immunosuppression and potential immunotherapeutic targets to convert Th-cell dysfunction.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zexin Zhang
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Bin Yin
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiayuan Li
- Department of Anesthesia Operation, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Cheng Xialin
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenqin Lian
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yingjun Su
- Department of Burns and Plastic Surgery, Plastic Surgery Hospital, Xi’an International Medical Center, Xi’an, Shaanxi, China
| | - Chiyu Jia
- Department of Burns and Plastic & Wound Repair Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
35
|
Isoform-selective HDAC Inhibitor Mocetinostat (MGCD0103) Alleviates Myocardial Ischemia/Reperfusion Injury via Mitochondrial Protection through the HDACs/CREB/PGC-1α Signaling Pathway. J Cardiovasc Pharmacol 2021; 79:217-228. [PMID: 34983914 DOI: 10.1097/fjc.0000000000001174] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/28/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Over the past decade, histone deacetylases (HDACs) has been proven to manipulate development and exacerbation of cardiovascular diseases, including myocardial ischemia/reperfusion injury (MIRI), cardiac hypertrophy, ventricular remodeling, myocardial fibrosis. Inhibition of histone deacetylases, especially class-I HDACs, is potent to protection of ischemic myocardium after ischemia/reperfusion. Herein, we examine whether mocetinostat (MGCD0103, MOCE), a class-I selective HDAC inhibitor in phase-II clinical trial, conducts cardioprotection under ischemia/reperfusion (I/R) in vivo and vitro, if so, reveal its potential pharmacological mechanism to provide an experimental and theoretical basis for mocetinostat usage in a clinical setting. HCMs were exposed to hypoxia and reoxygenation (H/R), with or without mocetinostat treatment. H/R reduced mitochondrial membrane potential (MMP) and induced HCMs apoptosis. Mocetinostat pre-treatment reversed these H/R-induced mitochondrial damage and cellular apoptosis and upregulated CREB, p-CREB and PGC-1α in HCMs during H/R. Transfection with siRNA against PGC-1α or CREB abolished the protective effects of mocetinostat on cardiomyocytes undergoing H/R. In vivo, mocetinostat was demonstrated to protect myocardial injury posed by myocardial ischemia/reperfusion (I/R) via activation of CREB and upregulation of PGC-1α. Mocetinostat (MGCD0103) can protect myocardium from ischemia/reperfusion injury through mitochondrial protection mediated by CREB/PGC-1α pathway. Therefore, activation of the CREB/PGC-1α signaling pathway via inhibition of Class-I HDACs may be a promising new therapeutic strategy for alleviating myocardial reperfusion injury.
Collapse
|
36
|
Zheng Z, Wang X, Wang Y, King JAC, Xie P, Wu S. CaMK4 is a downstream effector of the α 1G T-type calcium channel to determine the angiogenic potential of pulmonary microvascular endothelial cells. Am J Physiol Cell Physiol 2021; 321:C964-C977. [PMID: 34586897 DOI: 10.1152/ajpcell.00216.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/27/2021] [Indexed: 01/25/2023]
Abstract
Pulmonary microvascular endothelial cells (PMVECs) uniquely express an α1G-subtype of voltage-gated T-type Ca2+ channel. We have previously revealed that the α1G channel functions as a background Ca2+ entry pathway that is critical for the cell proliferation, migration, and angiogenic potential of PMVECs, a novel function attributed to the coupling between α1G-mediated Ca2+ entry and constitutive Akt phosphorylation and activation. Despite this significance, mechanism(s) that link the α1G-mediated Ca2+ entry to Akt phosphorylation remain incompletely understood. In this study, we demonstrate that Ca2+/calmodulin-dependent protein kinase (CaMK) 4 serves as a downstream effector of the α1G-mediated Ca2+ entry to promote the angiogenic potential of PMVECs. Notably, CaMK2 and CaMK4 are both expressed in PMVECs. Pharmacological blockade or genetic knockdown of the α1G channel led to a significant reduction in the phosphorylation level of CaMK4 but not the phosphorylation level of CaMK2. Pharmacological inhibition as well as genetic knockdown of CaMK4 significantly decreased cell proliferation, migration, and network formation capacity in PMVECs. However, CaMK4 inhibition or knockdown did not alter Akt phosphorylation status in PMVECs, indicating that α1G/Ca2+/CaMK4 is independent of the α1G/Ca2+/Akt pathway in sustaining the cells' angiogenic potential. Altogether, these findings suggest a novel α1G-CaMK4 signaling complex that regulates the Ca2+-dominated angiogenic potential in PMVECs.
Collapse
Affiliation(s)
- Zhen Zheng
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xuelin Wang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yuxia Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Judy A C King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, Louisiana
| | - Peilin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
37
|
Liu J, Li Y, Gao N, Ji J, He Q. Calcium/calmodulin-dependent protein kinase IV regulates vascular autophagy and insulin signaling through Akt/mTOR/CREB pathway in ob/ob mice. J Physiol Biochem 2021; 78:199-211. [PMID: 34741274 DOI: 10.1007/s13105-021-00853-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/20/2021] [Indexed: 10/19/2022]
Abstract
Calcium/calmodulin-dependent protein kinase IV (CaMKIV) has recently emerged as an important regulator of glucose metabolism and vascular function, but the underlying mechanism is not fully understood. Recently, we revealed that CaMKIV limits metabolic disorder and liver insulin resistance and regulates autophagy in high-fat diet-induced obese mice. In the present study, we demonstrated that CaMKIV was not only associated with improvement of glucose tolerance and insulin sensitivity in ob/ob mice but also involved in the regulation of vascular autophagy and mitochondrial biogenesis. Our in vitro data indicated that CaMKIV reversed autophagic imbalance and restored insulin sensitivity in palmitate-induced A7r5 cells with insulin resistance. However, the protective effects of CaMKIV were nullified by suppression of Akt, mTOR, or CREB, suggesting that CaMKIV inhibits autophagy and improves insulin signaling in insulin resistance cell models in an Akt/mTOR/CREB-dependent manner. CaMKIV reversed autophagic imbalance and insulin sensitivity in vascular tissues and vascular cells through Akt/mTOR/CREB signaling, which could be regarded as a novel opportunity for the treatment of insulin resistance.
Collapse
Affiliation(s)
- Jiali Liu
- Department of Clinical Laboratory, Xi'an Jiaotong University Second Affiliated Hospital, 157 West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Yue Li
- Department of Clinical Laboratory, Xi'an Jiaotong University Second Affiliated Hospital, 157 West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Ning Gao
- Department of Clinical Laboratory, Xi'an Jiaotong University Second Affiliated Hospital, 157 West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Jing Ji
- Department of Clinical Laboratory, Xi'an Jiaotong University Second Affiliated Hospital, 157 West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Qian He
- Department of Clinical Laboratory, Xi'an Jiaotong University Second Affiliated Hospital, 157 West 5 Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
38
|
Yang Y, Yu WW, Yan W, Xia Q. Decorin Induces Cardiac Hypertrophy by Regulating the CaMKII/MEF-2 Signaling Pathway In Vivo. Curr Med Sci 2021; 41:857-862. [PMID: 34643879 DOI: 10.1007/s11596-021-2426-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/20/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Cardiac hypertrophy is an adaptive reaction of the heart against cardiac overloading, but continuous cardiac hypertrophy can lead to cardiac remodeling and heart failure. Cardiac hypertrophy is mostly considered reversible, and recent studies have indicated that decorin not only prevents cardiac fibrosis associated with hypertension, but also achieves therapeutic effects by blocking fibrosis-related signaling pathways. However, the mechanism of action of decorin remains unknown and unconfirmed. METHODS We determined the degree of myocardial hypertrophy by measuring the ratios of the heart weight/body weight and left ventricular weight/body weight, histological analysis and immunohistochemistry. Western blotting was performed to detect the expression levels of CaMKII, p-CaMKII and MEF-2 in the heart. RESULTS Our results confirmed that decorin can regulate the CaMKII/MEF-2 signaling pathway, with inhibition thereof being similar to that of decorin in reducing cardiac hypertrophy. CONCLUSION Taken together, the results of the present study showed that decorin induced cardiac hypertrophy by regulating the CaMKII/MEF-2 signaling pathway in vivo, revealing a new therapeutic approach for the prevention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Yan Yang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei-Wei Yu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Xia
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
39
|
Yasuda K, Matsubara T, Shirakawa T, Kawamoto T, Kokabu S. Protein phosphatase 1 regulatory subunit 18 suppresses the transcriptional activity of NFATc1 via regulation of c-fos. Bone Rep 2021; 15:101114. [PMID: 34401407 PMCID: PMC8353383 DOI: 10.1016/j.bonr.2021.101114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/17/2021] [Accepted: 08/01/2021] [Indexed: 12/20/2022] Open
Abstract
The transcription factor NFATc1 and its binding partner AP-1 (a complex containing c-fos and c-Jun) play a central role in osteoclast differentiation. NFATc1 and AP-1 promote the expression of target genes such as Acp5, Ctsk and also auto-regulate NFATc1 expression as well. We previously reported that protein phosphatase 1 regulatory subunit 18 (PPP1r18) is a negative regulator of osteoclast bone resorption by inhibiting cell attachment to bone matrix. We also reported that PPP1r18 potentially regulates NFATc1 expression during osteoclast differentiation. To further explore this, in this study we have examined the effect of PPP1r18 on NFATc1 expression and activity by overexpressing PPP1r18 during the early stage of osteoclast differentiation. We found that PPP1r18 suppressed NFATc1 expression through inhibition of the transcriptional activity of NFATc1. Since PPP1r18 does not regulate NFATc1 directly, we next explored the involvement of AP-1. Our data showed that c-fos phosphorylation and nuclear localization were reduced by PPP1r18 overexpression. Further experiments showed that overexpression of c-fos together with PPP1r18 rescued NFATc1 expression and transcriptional activity. Moreover, c-fos activity inhibition by PPP1r18 was canceled by mutation of the phosphatase binding site of PPP1r18. Taken together, PPP1r18-regulated phosphatase activity targets c-fos phosphorylation and suppresses subsequent NFATc1 expression and activity.
PPP1r18 suppresses osteoclast differentiation. PPP1r18 suppresses c-fos phosphorylation and nuclear localization. PPP1r18 suppresses NFAT via c-fos.
Collapse
Key Words
- Ctsk, cathepsin K
- Dc-stamp, dendrocyte expressed seven transmembrane protein
- GapDH, glyceraldehyde-3-phosphate dehydrogenase
- M-CSF, macrophage colony stimulating factor
- NFATc1
- NFATc1, nuclear factor of activated T cells 1
- Osteoclast
- PP1, protein phosphatase 1
- PPP1r18
- PPP1r18, protein phosphatase 1 regulatory subunit 18
- RANK, receptor activator nuclear factor kappa B
- RANKL, receptor activator nuclear factor kappa B ligand
- Src, Rous sarcoma oncogene
- TRAP, tartrate resistant acid phosphatase
- c-Fos
- c-Jun, Jun proto-oncogene, AP-1 transcription factor subunit
- c-fos, Fos proto-oncogene, AP-1 transcription factor subunit
Collapse
Affiliation(s)
- Kazuma Yasuda
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Corresponding authors.
| | - Tomohiko Shirakawa
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Corresponding authors.
| |
Collapse
|
40
|
Kitazawa R, Haraguchi R, Kohara Y, Kitazawa S. RANK- NFATc1 signaling forms positive feedback loop on rank gene expression via functional NFATc1 responsive element in rank gene promoter. Biochem Biophys Res Commun 2021; 572:86-91. [PMID: 34358968 DOI: 10.1016/j.bbrc.2021.07.100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Receptor Activator of NF-κB (RANK) expressed on osteoclasts and their precursors is a receptor for RANK ligand (RANKL). Signals transduced by RANKL-RANK interaction induce genes essential for the differentiation and function of osteoclasts, partly through the direct binding of NFATc1, to target gene promoters. We have previously cloned a 6-kb fragment containing the 5'-flanking region of the mouse RANK gene and have demonstrated the presence of binding elements of hematological transcription factors, such as MITF, PU.1 and AP-1. Here, we demonstrated the presence of the functional NFATc1 responsive element on the RANK gene promoter. Transfection of an NFATc1-expression vector increased RANK mRNA that was subsequently nullified by NFATc1 knockdown. With the use of electrophoretic mobility shift assay (EMSA), an oligonucleotide (-388/-353) showed specific protein-DNA binding that was blockshifted with an anti-NFATc1 antibody and washed out with excess amounts of the cold consensus sequence. Co-transfection studies with the use of an NFATc1-expression vector and RANK promoter-reporter constructs showed that NFATc1 increased promoter activity 2-fold in RAW264.7 cells that was again nullified as disclosed by mutagenesis studies. Taken together, these results indicate that RANK transcription is positively regulated by the RANKL signal through the direct binding of NFATc1 to its specific binding site of the RANK gene promoter, and suggest the presence of a crucial positive feedback mechanism of gene expression that promotes accelerated terminal differentiation of RANK-positive committed precursors to mature osteoclasts.
Collapse
Affiliation(s)
- Riko Kitazawa
- Department of Molecular Pathology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan; Division of Diagnostic Molecular Pathology, Kobe University Graduate School of Medicine, Kusunoki-cho 7-5-1, Kobe, 650-0017, Japan; Division of Diagnostic Pathology, Ehime University Hospital, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Ryuma Haraguchi
- Department of Molecular Pathology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Yukihiro Kohara
- Department of Molecular Pathology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan
| | - Sohei Kitazawa
- Department of Molecular Pathology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon City, Ehime, 791-0295, Japan; Division of Diagnostic Molecular Pathology, Kobe University Graduate School of Medicine, Kusunoki-cho 7-5-1, Kobe, 650-0017, Japan.
| |
Collapse
|
41
|
Akizuki K, Ono A, Xue H, Kameshita I, Ishida A, Sueyoshi N. Biochemical characterization of four splice variants of mouse Ca2+/calmodulin-dependent protein kinase Iδ. J Biochem 2021; 169:445-458. [PMID: 33417706 DOI: 10.1093/jb/mvaa117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/14/2020] [Indexed: 11/12/2022] Open
Abstract
Ca2+/calmodulin (CaM)-dependent protein kinase Iδ (CaMKIδ) is a Ser/Thr kinase that plays pivotal roles in Ca2+ signalling. CaMKIδ is activated by Ca2+/CaM-binding and phosphorylation at Thr180 by CaMK kinase (CaMKK). In this study, we characterized four splice variants of mouse CaMKIδ (mCaMKIδs: a, b, c and d) found by in silico analysis. Recombinant mCaMKIδs expressed in Escherichia coli were phosphorylated by CaMKK; however, only mCaMKIδ-a and c showed protein kinase activities towards myelin basic protein in vitro, with mCaMKIδ-b and mCaMKIδ-d being inactive. Although mCaMKIδ-a and mCaMKIδ-c underwent autophosphorylation in vitro, only mCaMKIδ-c underwent autophosphorylation in 293T cells. Site-directed mutagenesis showed that the autophosphorylation site is Ser349, which is found in the C-terminal region of only variants c and b (Ser324). Furthermore, phosphorylation of these sites (Ser324 and Ser349) in mCaMKIδ-b and c was more efficiently catalyzed by cAMP-dependent protein kinase in vitro and in cellulo as compared to the autophosphorylation of mCaMKIδ-c. Thus, variants of mCaMKIδ possess distinct properties in terms of kinase activities, autophosphorylation and phosphorylation by another kinase, suggesting that they play physiologically different roles in murine cells.
Collapse
Affiliation(s)
- Kazutoshi Akizuki
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, 2393 Ikenobe, Miki, Kagawa 761-0795, Japan.,Research Fellow of Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan.,Laboratory of Molecular Brain Science, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8521, Japan
| | - Ayaka Ono
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, 2393 Ikenobe, Miki, Kagawa 761-0795, Japan
| | - Houcheng Xue
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, 2393 Ikenobe, Miki, Kagawa 761-0795, Japan
| | - Isamu Kameshita
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, 2393 Ikenobe, Miki, Kagawa 761-0795, Japan
| | - Atsuhiko Ishida
- Laboratory of Molecular Brain Science, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8521, Japan
| | - Noriyuki Sueyoshi
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, 2393 Ikenobe, Miki, Kagawa 761-0795, Japan
| |
Collapse
|
42
|
Kim HJ, Lee J, Lee GR, Kim N, Lee HI, Kwon M, Kim NY, Park JH, Kang YH, Song HJ, Kim T, Shin DM, Jeong W. Flunarizine inhibits osteoclastogenesis by regulating calcium signaling and promotes osteogenesis. J Cell Physiol 2021; 236:8239-8252. [PMID: 34192358 DOI: 10.1002/jcp.30496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 11/12/2022]
Abstract
Many bone diseases such as osteoporosis and periodontitis are caused by hyperactivation of osteoclasts. Calcium (Ca2+ ) signals are crucial for osteoclast differentiation and function. Thus, the blockade of Ca2+ signaling may be a strategy for regulating osteoclast activity and has clinical implications. Flunarizine (FN) is a Ca2+ channel antagonist that has been used for reducing migraines. However, the role of FN in osteoclast differentiation and function remains unknown. Here, we investigated whether FN regulates osteoclastogenesis and elucidated the molecular mechanism. FN inhibited osteoclast differentiation along with decreased expression of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), and attenuated osteoclast maturation and bone resorption. FN inhibition of osteoclast differentiation was restored by ectopic expression of constitutively active NFATc1. FN reduced calcium oscillations and its inhibition of osteoclast differentiation and resorption function was reversed by ionomycin, an ionophore that binds Ca2+ . FN also inhibited Ca2+ /calmodulin-dependent protein kinase IV (CaMKIV) and calcineurin leading to a decrease in the cAMP-responsive element-binding protein-dependent cFos and peroxisome proliferator-activated receptor-γ coactivator 1β expression, and NFATc1 nuclear translocation. These results indicate that FN inhibits osteoclastogenesis via regulating CaMKIV and calcineurin as a Ca2+ channel blocker. In addition, FN-induced apoptosis in osteoclasts and promoted osteogenesis. Furthermore, FN protected lipopolysaccharide- and ovariectomy-induced bone destruction in mouse models, suggesting that it has therapeutic potential for treating inflammatory bone diseases and postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Jiae Lee
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Gong-Rak Lee
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Narae Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Hye In Lee
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Minjeong Kwon
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Nam Young Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Jin Ha Park
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Ye Hee Kang
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Hyeong Ju Song
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - TaeSoo Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Dong Min Shin
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - Woojin Jeong
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| |
Collapse
|
43
|
Abstract
PURPOSE Down syndrome (DS) is caused by trisomy 21 (Ts21) and results in skeletal deficits including shortened stature, low bone mineral density, and a predisposition to early onset osteoporosis. Ts21 causes significant alterations in skeletal development, morphology of the appendicular skeleton, bone homeostasis, age-related bone loss, and bone strength. However, the genetic or cellular origins of DS skeletal phenotypes remain unclear. RECENT FINDINGS New studies reveal a sexual dimorphism in characteristics and onset of skeletal deficits that differ between DS and typically developing individuals. Age-related bone loss occurs earlier in the DS as compared to general population. Perturbations of DS skeletal quality arise from alterations in cellular and molecular pathways affected by the overexpression of trisomic genes. Sex-specific alterations occur in critical developmental pathways that disrupt bone accrual, remodeling, and homeostasis and are compounded by aging, resulting in increased risks for osteopenia, osteoporosis, and fracture in individuals with DS.
Collapse
Affiliation(s)
- Jared R Thomas
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL 306, Indianapolis, IN, 46202-3275, USA
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL 306, Indianapolis, IN, 46202-3275, USA.
| |
Collapse
|
44
|
Yokota K, Sato K, Miyazaki T, Aizaki Y, Tanaka S, Sekikawa M, Kozu N, Kadono Y, Oda H, Mimura T. Characterization and Function of Tumor Necrosis Factor and Interleukin-6-Induced Osteoclasts in Rheumatoid Arthritis. Arthritis Rheumatol 2021; 73:1145-1154. [PMID: 33512089 PMCID: PMC8361923 DOI: 10.1002/art.41666] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 01/21/2021] [Indexed: 12/18/2022]
Abstract
Objective We have previously reported that stimulation of mouse bone marrow–derived macrophages with tumor necrosis factor (TNF) and interleukin‐6 (IL‐6) induces differentiation of osteoclast‐like cells. We undertook this study to clarify the characterization and function of human TNF and IL‐6–induced osteoclasts using peripheral blood collected from patients with rheumatoid arthritis (RA) and healthy donors. Methods Peripheral blood monocytes were cultured with a combination of TNF and IL‐6, TNF alone, IL‐6 alone, or with RANKL, and their bone resorption ability was evaluated. Expression levels of NFATc1, proinflammatory cytokines, and matrix metalloproteinase 3 were analyzed. The effects of NFAT inhibitor and JAK inhibitor were examined. Furthermore, the relationship between the number of TNF and IL‐6–induced osteoclasts or RANKL‐induced osteoclasts differentiated from peripheral blood mononuclear cells (PBMCs) in patients with RA and the modified total Sharp score (mTSS) or whole‐body bone mineral density (BMD) was examined. Results Peripheral blood monocytes stimulated with a TNF and IL‐6–induced osteoclasts were shown to demonstrate the ability to absorb bone matrix. Cell differentiation was not inhibited by the addition of osteoprotegerin. Stimulation with a combination of TNF and IL‐6 promoted NFATc1 expression, whereas the NFAT and JAK inhibitors prevented TNF and IL‐6–induced osteoclast formation. Expression levels of IL1β, TNF, IL12p40, and MMP3 were significantly increased in TNF and IL‐6–induced osteoclasts, but not in RANKL‐induced osteoclasts. The number of TNF and IL‐6–induced osteoclasts differentiated from PBMCs in patients with RA positively correlated with the mTSS, whereas RANKL‐induced osteoclast numbers negatively correlated with the whole‐body BMD of the same patients. Conclusion Our results demonstrate that TNF and IL‐6–induced osteoclasts may contribute to the pathology of inflammatory arthritis associated with joint destruction, such as RA.
Collapse
Affiliation(s)
- Kazuhiro Yokota
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| | - Kojiro Sato
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | | | - Yoshimi Aizaki
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| | - Shinya Tanaka
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | - Miyoko Sekikawa
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | | | - Yuho Kadono
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| | - Hiromi Oda
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | - Toshihide Mimura
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| |
Collapse
|
45
|
Elango J, Bao B, Wu W. The hidden secrets of soluble RANKL in bone biology. Cytokine 2021; 144:155559. [PMID: 33994070 DOI: 10.1016/j.cyto.2021.155559] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/25/2022]
Abstract
The discovery of cytokine tumor necrosis factor (TNF) in the 20th century revealed numerous secrets about organ development. In particular, the functions identified for the receptor activator of nuclear factor kappa-β (NF-κβ) ligand (also known as the RANKL/osteoprotegerin ligand (OPGL) or RANK ligand/TNFSF11) in the homeostasis of skeletal structure, function and regulation were not anticipated. Empirical evidence established the receptor-ligand interaction of RANKL with RANK in osteoclast formation. Reverse signaling of RANKL triggers NF-κβ for the degradation of β-catenin to inhibit bone formation. There is also evidence that RANKL modifies the behavior of other cells in the bone microenvironment, including osteoblasts, chondrocytes, endothelial cells and lymphocytes during normal (homeostatic) and diseased (osteoimmune) states. Two forms of RANKL, i.e., soluble and membrane-bound RANKL, are produced by bone cells. Even though soluble RANKL (sRANKL) and membrane-bound RANKL (mRANKL) both stimulate osteoclast formation in vitro, their biological roles are different. mRANKL triggers osteoclastogenesis by binding to RANK through cell-cell interaction; however, sRANKL released from osteogenic cells binds to RANK without cell-cell interaction. This review attempts to hypothesize how sRANKL functions biologically in bone and explore how this hypothesis might influence future research.
Collapse
Affiliation(s)
- Jeevithan Elango
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - Bin Bao
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Wenhui Wu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
46
|
The Calcium/Calmodulin-Dependent Kinases II and IV as Therapeutic Targets in Neurodegenerative and Neuropsychiatric Disorders. Int J Mol Sci 2021; 22:ijms22094307. [PMID: 33919163 PMCID: PMC8122486 DOI: 10.3390/ijms22094307] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
CaMKII and CaMKIV are calcium/calmodulin-dependent kinases playing a rudimentary role in many regulatory processes in the organism. These kinases attract increasing interest due to their involvement primarily in memory and plasticity and various cellular functions. Although CaMKII and CaMKIV are mostly recognized as the important cogs in a memory machine, little is known about their effect on mood and role in neuropsychiatric diseases etiology. Here, we aimed to review the structure and functions of CaMKII and CaMKIV, as well as how these kinases modulate the animals’ behavior to promote antidepressant-like, anxiolytic-like, and procognitive effects. The review will help in the understanding of the roles of the above kinases in the selected neurodegenerative and neuropsychiatric disorders, and this knowledge can be used in future drug design.
Collapse
|
47
|
Nedeva IR, Vitale M, Elson A, Hoyland JA, Bella J. Role of OSCAR Signaling in Osteoclastogenesis and Bone Disease. Front Cell Dev Biol 2021; 9:641162. [PMID: 33912557 PMCID: PMC8072347 DOI: 10.3389/fcell.2021.641162] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Formation of mature bone-resorbing cells through osteoclastogenesis is required for the continuous remodeling and repair of bone tissue. In aging and disease this process may become aberrant, resulting in excessive bone degradation and fragility fractures. Interaction of receptor-activator of nuclear factor-κB (RANK) with its ligand RANKL activates the main signaling pathway for osteoclastogenesis. However, compelling evidence indicates that this pathway may not be sufficient for the production of mature osteoclast cells and that co-stimulatory signals may be required for both the expression of osteoclast-specific genes and the activation of osteoclasts. Osteoclast-associated receptor (OSCAR), a regulator of osteoclast differentiation, provides one such co-stimulatory pathway. This review summarizes our present knowledge of osteoclastogenesis signaling and the role of OSCAR in the normal production of bone-resorbing cells and in bone disease. Understanding the signaling mechanism through this receptor and how it contributes to the production of mature osteoclasts may offer a more specific and targeted approach for pharmacological intervention against pathological bone resorption.
Collapse
Affiliation(s)
- Iva R Nedeva
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Mattia Vitale
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Jordi Bella
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
48
|
Wang Y, Zheng Y, Li W. Compression loading of osteoclasts attenuated microRNA-146a-5p expression, which promotes angiogenesis by targeting adiponectin. SCIENCE CHINA-LIFE SCIENCES 2021; 65:151-166. [PMID: 33677819 DOI: 10.1007/s11427-020-1869-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/06/2021] [Indexed: 11/24/2022]
Abstract
Osteoclastogenesis in alveolar bone induced by compression stress triggers orthodontic tooth movement. Compression stress also stimulates angiogenesis, which is essential for osteoclastogenesis. However, the effects of osteoclastogenesis induced by compression on angiogenesis are poorly understood. In vivo, we found the markers of angiogenesis increased during orthodontic bone remodeling. In vitro, osteoclast-derived exosomes increased proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs), as well as expression of vascular endothelial growth factor and CD31. The promotive effects of exosomes derived from compressed osteoclasts were greater than those derived from osteoclasts without compression. Next, we analyzed changes in the microRNA transcriptome after compression stress and focused on microRNA146a-5p (miR-146a), which was significantly decreased by compression. Transfection of an inhibitor of miR-146a stimulated angiogenesis of HUVECs while miR-146a mimics repressed angiogenesis. Adiponectin (ADP) was confirmed to be a target of miR-146a by dual luciferase reporter assay. In HUVECs treated with exosomes, we detected increased ADP which promoted angiogenesis. Knockdown of ADP in HUVECs reduced the promotive effects of exosomes. Our results demonstrate that the decreased miR-146a observed in osteoclasts after compression promotes angiogenesis by targeting ADP, suggesting a novel method to interfere with bone remodeling induced by compression stress.
Collapse
Affiliation(s)
- Yue Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
49
|
Li Y, Lin S, Liu P, Huang J, Qiu J, Wen Z, Yuan J, Qiu H, Liu Y, Liu Q, Zhou T, Luo P, Guo H, Ma Y, Guo D, Mo G, Tang Y, Xu L, Liang D, Xu J, Ding Y, Zhang S. Carnosol suppresses RANKL-induced osteoclastogenesis and attenuates titanium particles-induced osteolysis. J Cell Physiol 2021; 236:1950-1966. [PMID: 32722851 DOI: 10.1002/jcp.29978] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022]
Abstract
Osteolysis is a common medical condition characterized by excessive activity of osteoclasts and bone resorption, leading to severe poor quality of life. It is essential to identify the medications that can effectively suppress the excessive differentiation and function of osteoclasts to prevent and reduce the osteolytic conditions. It has been reported that Carnosol (Car), isolated from rosemary and salvia, has anti-inflammatory, antioxidative, and anticancer effects, but its activity on osteolysis has not been determined. In this study, we found that Car has a strong inhibitory effect on the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation dose-dependently without any observable cytotoxicity. Moreover, Car can inhibit the RANKL-induced osteoclastogenesis and resorptive function via suppressing NFATc1, which is a result of affecting MAPK, NF-κB and Ca2+ signaling pathways. Moreover, the particle-induced osteolysis mouse model confirmed that Car could be effective for the treatment of bone loss in vivo. Taken together, by suppressing the formation and function of RANKL-induced osteoclast, Car, may be a therapeutic supplementary in the prevention or the treatment of osteolysis.
Collapse
Affiliation(s)
- Yongxian Li
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Sipeng Lin
- Orthopaedic Department, Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Panjie Liu
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianbin Huang
- Orthopaedic Department, Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Junxiong Qiu
- Orthopaedic Department, Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhenkang Wen
- Orthopaedic Department, Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jinbo Yuan
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Heng Qiu
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Yuhao Liu
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Tengpeng Zhou
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peijie Luo
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huizhi Guo
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhuai Ma
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Danqing Guo
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoye Mo
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongchao Tang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangliang Xu
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Yue Ding
- Orthopaedic Department, Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuncong Zhang
- The First Clinical Academy, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
50
|
Jo YJ, Lee HI, Kim N, Hwang D, Lee J, Lee GR, Hong SE, Lee H, Kwon M, Kim NY, Kim HJ, Park JH, Kang YH, Kim HS, Lee SY, Jeong W. Cinchonine inhibits osteoclast differentiation by regulating TAK1 and AKT, and promotes osteogenesis. J Cell Physiol 2021; 236:1854-1865. [PMID: 32700766 DOI: 10.1002/jcp.29968] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/26/2020] [Accepted: 07/14/2020] [Indexed: 01/09/2023]
Abstract
Cinchonine (CN) has been known to exert antimalarial, antiplatelet, and antiobesity effects. It was also recently reported to inhibit transforming growth factor β-activated kinase 1 (TAK1) and protein kinase B (AKT) through binding to tumor necrosis factor receptor-associated factor 6 (TRAF6). However, its role in bone metabolism remains largely unknown. Here, we showed that CN inhibits osteoclast differentiation with decreased expression of nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), a key determinant of osteoclastogenesis. Immunoblot and quantitative real-time polymerase chain reaction analysis as well as the reporter assay revealed that CN inhibits nuclear factor-κB and activator protein-1 by regulating TAK1. CN also attenuated the activation of AKT, cyclic AMP response element-binding protein, and peroxisome proliferator-activated receptor-γ coactivator 1β (PGC1β), an essential regulator of mitochondrial biogenesis. Collectively, these results suggested that CN may inhibit TRAF6-mediated TAK1 and AKT activation, which leads to downregulation of NFATc1 and PGC1β resulting in the suppression of osteoclast differentiation. Interestingly, CN not only inhibited the maturation and resorption function of differentiated osteoclasts but also promoted osteoblast differentiation. Furthermore, CN protected lipopolysaccharide- and ovariectomy-induced bone destruction in mouse models, suggesting its therapeutic potential for treating inflammation-induced bone diseases and postmenopausal osteoporosis.
Collapse
Affiliation(s)
- You-Jin Jo
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Hye In Lee
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Narae Kim
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Donghyun Hwang
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea
| | - Jiae Lee
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Gong-Rak Lee
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Seong-Eun Hong
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea
| | - Minjeong Kwon
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Nam Young Kim
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Hyun Jin Kim
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Jin Ha Park
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Ye Hee Kang
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, Republic of Korea
| | - Soo Young Lee
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| | - Woojin Jeong
- Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, South Korea
| |
Collapse
|