1
|
Lynskey SJ, Gill SD, McGee SL, Ziemann M, Page RS. 'QuickDASH' to find unique genes and biological processes associated with shoulder osteoarthritis: a prospective case-control study. BMC Res Notes 2024; 17:361. [PMID: 39702481 DOI: 10.1186/s13104-024-07035-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a disease impacting the synovial joint complex, yet transcriptional changes specific to shoulder OA remain underexplored. This study aims to profile transcriptomic changes in periarticular tissues from patients undergoing shoulder replacement for OA. By correlating these profiles with QuickDASH scores-a validated measure of worsening shoulder function-this research seeks to understand the gene expression changes associated with clinical decline. Capsular tissue biopsies from shoulder OA patients were compared with those from a control group undergoing shoulder stabilization for recurrent instability. This investigation forms part of a larger transcriptomic analysis of painful shoulder conditions which will address the current gap in knowledge regarding the molecular and genetic underpinnings of shoulder OA, rotator cuff tears and cuff-tear arthropathy. RESULTS The analysis revealed that genes most strongly associated with increasing QuickDASH scores across tissues were linked to inflammation and stress response. Key pathways involved interleukins, chemokines, complement components, nuclear response factors, and immediate early response genes, reflecting a balance between pro- and anti-inflammatory signalling. Additionally, this study identified unique gene expression patterns in shoulder OA not previously observed in hip and knee OA, along with novel genes implicated in shoulder OA, highlighting areas for future targeted investigation. Trial registration This investigation has been registered with the Australian New Zealand Clinical Trials Registry (ANZCTR), registered on the 26th of March 2018, registration number: 12618000431224, accessible from: https://anzctr.org.au/Trial/Registration/TrialReview.aspx?id=374665&isReview=true.
Collapse
Affiliation(s)
- Samuel J Lynskey
- Department of Orthopaedic Surgery, Geelong University Hospital, Geelong, Australia.
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong, VIC, Australia.
| | - Stephen D Gill
- Department of Orthopaedic Surgery, Geelong University Hospital, Geelong, Australia
- Barwon Centre for Orthopaedic Research and Education (BCORE), St John of God Hospital, Deakin University, Geelong, Australia
- Geelong University Hospital, Geelong, Australia
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Sean L McGee
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong, VIC, Australia
| | - Mark Ziemann
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong, VIC, Australia
| | - Richard S Page
- Department of Orthopaedic Surgery, Geelong University Hospital, Geelong, Australia
- Barwon Centre for Orthopaedic Research and Education (BCORE), St John of God Hospital, Deakin University, Geelong, Australia
- Geelong University Hospital, Geelong, Australia
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
2
|
Tian K, He X, Lin X, Chen X, Su Y, Lu Z, Chen Z, Zhang L, Li P, Ma L, Lan Z, Zhao X, Fen G, Hai Q, Xue D, Jin Q. Unveiling the Role of Sik1 in Osteoblast Differentiation: Implications for Osteoarthritis. Mol Cell Biol 2024; 44:411-428. [PMID: 39169784 PMCID: PMC11485870 DOI: 10.1080/10985549.2024.2385633] [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: 03/25/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by subchondral osteosclerosis, mainly due to osteoblast activity. This research investigates the function of Sik1, a member of the AMP-activated protein kinase family, in OA. Proteomic analysis was conducted on clinical samples from 30 OA patients, revealing a negative correlation between Sik1 expression and OA. In vitro experiments utilized BMSCs to examine the effect of Sik1 on osteogenic differentiation. BMSCs were cultured and induced toward osteogenesis with specific media. Sik1 overexpression was achieved through lentiviral transfection, followed by analysis of osteogenesis-associated proteins using Western blotting, RT-qPCR, and alkaline phosphate staining. In vivo experiments involved destabilizing the medial meniscus in mice to establish an OA model, assessing the therapeutic potential of Sik1. The CT scans and histological staining were used to analyze subchondral bone alterations and cartilage damage. The findings show that Sik1 downregulation correlates with advanced OA and heightened osteogenic differentiation in BMSCs. Sik1 overexpression inhibits osteogenesis-related markers in vitro and reduces cartilage damage and subchondral osteosclerosis in vivo. Mechanistically, Sik1 modulates osteogenesis and subchondral bone changes through Runx2 activity regulation. The research emphasizes Sik1 as a promising target for treating OA, suggesting its involvement in controlling bone formation and changes in the subchondral osteosclerosis.
Collapse
Affiliation(s)
- Kuanmin Tian
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaoxin He
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xue Lin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaolei Chen
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yajing Su
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhidong Lu
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhirong Chen
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Liang Zhang
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Peng Li
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Long Ma
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhibin Lan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xin Zhao
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Gangning Fen
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qinqin Hai
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Di Xue
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qunhua Jin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| |
Collapse
|
3
|
Mo S, Kim MK, Jang JS, Lee SH, Hong SJ, Jung S, Kim HH. Unique expression and critical role of metallothionein 3 in the control of osteoclastogenesis and osteoporosis. Exp Mol Med 2024; 56:1791-1806. [PMID: 39085359 PMCID: PMC11372110 DOI: 10.1038/s12276-024-01290-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 08/02/2024] Open
Abstract
Bone homeostasis is maintained by an intricate balance between osteoclasts and osteoblasts, which becomes disturbed in osteoporosis. Metallothioneins (MTs) are major contributors in cellular zinc regulation. However, the role of MTs in bone cell regulation has remained unexplored. Single-cell RNA sequencing analysis discovered that, unlike the expression of other MT members, the expression of MT3 was unique to osteoclasts among various macrophage populations and was highly upregulated during osteoclast differentiation. This unique MT3 upregulation was validated experimentally and supported by ATAC sequencing data analyses. Downregulation of MT3 by gene knockdown or knockout resulted in excessive osteoclastogenesis and exacerbated bone loss in ovariectomy-induced osteoporosis. Transcriptome sequencing of MT3 knockdown osteoclasts and gene set enrichment analysis indicated that the oxidative stress and redox pathways were enriched, which was verified by MT3-dependent regulation of reactive oxygen species (ROS). In addition, MT3 deficiency increased the transcriptional activity of SP1 in a manner dependent on intracellular zinc levels. This MT3-zinc-SP1 axis was crucial for the control of osteoclasts, as zinc chelation and SP1 knockdown abrogated the promotion of SP1 activity and osteoclastogenesis by MT3 deletion. Moreover, SP1 bound to the NFATc1 promoter, and overexpression of an inactive SP1 mutant negated the effects of MT3 deletion on NFATc1 and osteoclastogenesis. In conclusion, MT3 plays a pivotal role in controlling osteoclastogenesis and bone metabolism via dual axes involving ROS and SP1. The present study demonstrated that MT3 elevation is a potential therapeutic strategy for osteolytic bone disorders, and it established for the first time that MT3 is a crucial bone mass regulator.
Collapse
Affiliation(s)
- Shenzheng Mo
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Bone Science R&D Center, Tissue Regeneration Institute, Osstem Implant, Seoul, 07789, Republic of Korea
| | - Ji Sun Jang
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seung Hye Lee
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seo Jin Hong
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Suhan Jung
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
4
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [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: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
5
|
Zhu YS, Yan H, Mo TT, Zhang JN, Jiang C. Identification of Diagnostic Markers in Synovial Tissue of Osteoarthritis by Weighted Gene Coexpression Network. Biochem Genet 2023; 61:2056-2075. [PMID: 36929359 DOI: 10.1007/s10528-023-10359-z] [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: 09/29/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
Osteoarthritis (OA) is a serious threat to human health. However, the etiology and pathogenesis of the disease are not fully understood. Most researchers believe that the degeneration and imbalance of articular cartilage, extracellular matrix, and subchondral bone are the fundamental causes of osteoarthritis. However, recent studies have shown that synovial lesions may precede cartilage, which may be an important precipitating factor in the early stage of OA and the whole course of the disease. This study aimed to conduct an analysis based on sequence data from the Gene Expression Omnibus (GEO) database to investigate the presence of effective biomarkers in the synovial tissue of osteoarthritis for the diagnosis and control of OA progression. In this study, the differentially expressed OA-related genes (DE-OARGs) in osteoarthritis synovial tissues were extracted in the GSE55235 and GSE55457 datasets using the Weighted Gene Co-expression Network Analysis (WGCNA) and limma. Least-Absolute Shrinkage and Selection Operator (LASSO) algorithm was used to select the diagnostic genes based on the DE-OARGs by glmnet package. 7 genes were selected as diagnostic genes including SAT1, RLF, MAFF, SIK1, RORA, ZNF529, and EBF2. Subsequently, the diagnostic model was constructed and the results of the Area Under the Curve (AUC) demonstrated that the diagnostic model had high diagnostic performance for OA. Additionally, among the 22 immune cells of the Cell type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) and the 24 immune cells of the single sample Gene Set Enrichment Analysis (ssGSEA), 3 immune cells and 5 immune cells were different between the OA and normal samples, respectively. The expression trends of the 7 diagnostic genes were consistent in the GEO datasets and the results of the real-time reverse transcription PCR (qRT-PCR). The results of this study demonstrate that these diagnostic markers have important significance in the diagnosis and treatment of OA, and will provide further evidence for the clinical and functional studies of OA.
Collapse
Affiliation(s)
- Yun-Sen Zhu
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Wenzhou Medical University Affiliated Wenling Hospital, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Hong Yan
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Wenzhou Medical University Affiliated Wenling Hospital, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Ting-Ting Mo
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Wenzhou Medical University Affiliated Wenling Hospital, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Jiang-Nan Zhang
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Wenzhou Medical University Affiliated Wenling Hospital, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China
| | - Chang Jiang
- Department of Orthopaedic Surgery, The First People's Hospital of Wenling, Wenzhou Medical University Affiliated Wenling Hospital, Chuan'an Nan Road NO 333, Wenling, 317500, Zhejiang, China.
| |
Collapse
|
6
|
Zhu J, Li C, Wang P, Liu Y, Li Z, Chen Z, Zhang Y, Wang B, Li X, Yan Z, Liang X, Zhou S, Ao X, Zhu M, Zhou P, Gu Y. Deficiency of salt-inducible kinase 2 (SIK2) promotes immune injury by inhibiting the maturation of lymphocytes. MedComm (Beijing) 2023; 4:e366. [PMID: 37706195 PMCID: PMC10495731 DOI: 10.1002/mco2.366] [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: 08/22/2022] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023] Open
Abstract
Salt-inducible kinase 2 (SIK2) belongs to the serine/threonine protein kinases of the AMPK/SNF1 family, which has important roles in cell cycle, tumor, melanogenesis, neuronal damage repair and apoptosis. Recent studies showed that SIK2 regulates the macrophage polarization to make a balance between inflammation and macrophage. Macrophage is critical to initiate immune regulation, however, whether SIK2 can be involved in immune regulation is not still well understood. Here, we revealed that the protein of SIK2 was highly expressed in thymus, spleen, lung, and brain. And SIK2 protein content increased in RAW264.7 and AHH1 cells with a time and dose-dependent after-ionizing radiation (IR). Inhibition of SIK2 could promote AHH1 cells apoptosis Moreover, we used the Cre-LoxP system to construct the SIK2+/- mice, and the research on function suggested that the deficiency of SIK2 could promote the sensitivity of IR. The deficiency of SIK2 promoted the immune injury via inhibiting the maturation of T cells and B cells. Furthermore, the TCRβ rearrangement was inhibited by the deficiency of SIK2. Collectively, this study demonstrated that SIK2 provides an essential function of regulating immune injury, which will provide new ideas for the treatment of immune injury-related diseases.
Collapse
Affiliation(s)
- Jiaojiao Zhu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
| | - Chao Li
- School of Life ScienceShihezi University, ShiheziXinjiang ProvinceP. R. China
| | - Ping Wang
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
| | - Yuhao Liu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
| | - Zhongqiu Li
- Medical SchoolShihezi University, ShiheziXinjiang ProvinceP. R. China
| | - Zhongmin Chen
- PLA Rocket Force Characteristic Medical CenterBeijingP. R. China
| | - Ying Zhang
- Medical SchoolShihezi University, ShiheziXinjiang ProvinceP. R. China
| | - Bin Wang
- School of Life ScienceShihezi University, ShiheziXinjiang ProvinceP. R. China
| | - Xueping Li
- School of Life ScienceShihezi University, ShiheziXinjiang ProvinceP. R. China
| | - Ziyan Yan
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
| | - Xinxin Liang
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceP. R. China
| | - Shenghui Zhou
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceP. R. China
| | - Xingkun Ao
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceP. R. China
| | - Maoxiang Zhu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
| | - Pingkun Zhou
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
| | - Yongqing Gu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingP. R. China
- Medical SchoolShihezi University, ShiheziXinjiang ProvinceP. R. China
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunan ProvinceP. R. China
| |
Collapse
|
7
|
Cai X, Wang L, Yi Y, Deng D, Shi M, Tang M, Li N, Wei H, Zhang R, Su K, Ye H, Chen L. Discovery of pyrimidine-5-carboxamide derivatives as novel salt-inducible kinases (SIKs) inhibitors for inflammatory bowel disease (IBD) treatment. Eur J Med Chem 2023; 256:115469. [PMID: 37178481 DOI: 10.1016/j.ejmech.2023.115469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Salt-inducible kinases (SIKs) play a crucial role in inflammation process, acting as molecular switches that regulate the transformation of M1/M2 macrophages. HG-9-91-01 is a SIKs inhibitor with potent inhibitory activity against SIKs in the nanomolar range. However, its poor drug-like properties, including a rapid elimination rate, low in vivo exposure and high plasma protein binding rate, have hindered further research and clinical application. To improve the drug-like properties of HG-9-91-01, a series of pyrimidine-5-carboxamide derivatives were designed and synthesized through a molecular hybridization strategy. The most promising compound 8h was obtained with favorable activity and selectivity on SIK1/2, excellent metabolic stability in human liver microsome, enhanced in vivo exposure and suitable plasma protein binding rate. Mechanism research showed that compound 8h significantly up-regulated the expression of anti-inflammatory cytokine IL-10 and reduced the expression of pro-inflammatory cytokine IL-12 in bone marrow-derived macrophages. Furthermore, it significantly elevated expression of cAMP response element-binding protein (CREB) target genes IL-10, c-FOS and Nurr77. Compound 8h also induced the translocation of CREB-regulated transcriptional coactivator 3 (CRTC3) and elevated the expression of LIGHT, SPHK1 and Arginase 1. Additionally, compound 8h demonstrated excellent anti-inflammatory effects in a DSS-induced colitis model. Generally, this research indicated that compound 8h has the potential to be developed as an anti-inflammatory drug candidate.
Collapse
Affiliation(s)
- Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lun Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyao Yi
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Dexin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingsong Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haoche Wei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Chengdu Zenitar Biomedical Technology Co., Ltd, Chengdu, China.
| |
Collapse
|
8
|
Study on the Mechanism of miR-146a in Gingival Mesenchymal Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1630260. [PMID: 36425259 PMCID: PMC9681548 DOI: 10.1155/2022/1630260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022]
Abstract
This study aimed to investigate the molecular mechanisms of microRNA-146a (miR-146a) on gingival mesenchymal stem cells (MSCs). Gingival MSCs were isolated from the gingiva tissues of patients with periodontal disease to reveal the function of miR-146a in regulating osteoblast differentiation. miR-146a inhibits osteoblast differentiation by inhibiting phosphorylated cyclic-AMP response binding (CREB) protein translocation into the nucleus and ultimately attenuating runt-related transcription factor 2 (Runx2) expression. Furthermore, silencing miR-146a promotes the proliferation of gingival MSCs. Of note, targeted inhibition of miR-146a also inhibited LPS-induced inflammatory response and promoted the proliferation of gingival MSCs via CREB/Runx2 axis. MiR-146a is a key negative regulator of gingival MSCs proliferation and osteogenic differentiation, and targeting to reduce the miR-146a expression is essential for bone formation signaling. Therefore, we propose that miR-146a is a useful therapeutic target for the development of bone anabolic strategies.
Collapse
|
9
|
Yoon SH, Tang CC, Wein MN. Salt inducible kinases and PTH1R action. VITAMINS AND HORMONES 2022; 120:23-45. [PMID: 35953111 DOI: 10.1016/bs.vh.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Parathyroid hormone is a central regulator of calcium homeostasis. PTH protects the organism from hypocalcemia through its actions in bone and kidney. Recent physiologic studies have revealed key target genes for PTH receptor (PTH1R) signaling in these target organs. However, the complete signal transduction cascade used by PTH1R to accomplish these physiologic actions has remained poorly defined. Here we will review recent studies that have defined an important role for salt inducible kinases downstream of PTH1R in bone, cartilage, and kidney. PTH1R signaling inhibits the activity of salt inducible kinases. Therefore, direct SIK inhibitors represent a promising novel strategy to mimic PTH actions using small molecules. Moreover, a detailed understanding of the molecular circuitry used by PTH1R to exert its biologic effects will afford powerful new models to better understand the diverse actions of this important G protein coupled receptor in health and disease.
Collapse
Affiliation(s)
- Sung-Hee Yoon
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Cheng-Chia Tang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
10
|
Xu H, Tong G, Yan T, Dong L, Yang X, Dou D, Sun Z, Liu T, Zheng X, Yang J, Sun X, Zhou Y, Kuang Y. Transcriptomic Analysis Provides Insights to Reveal the bmp6 Function Related to the Development of Intermuscular Bones in Zebrafish. Front Cell Dev Biol 2022; 10:821471. [PMID: 35646941 PMCID: PMC9135397 DOI: 10.3389/fcell.2022.821471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
Intermuscular bones (IBs) are small, hard-boned spicules located in the muscle tissue that mainly exist in the myosepta of lower teleosts, which hurt the edibleness and economic value of fish. The study of the development of IBs is very important for freshwater aquaculture fish, but the molecular mechanism of its formation and the key regulatory genes remain unclear. In this study, we first constructed two types of zebrafish mutants (the mutants losing IBs and the mutants with partial deletion of IBs) by knocking out bmp6. We then carried out a transcriptomic analysis to reveal the role of bmp6 in the developmental mechanism of IBs; we used the caudal musculoskeletal tissues of these mutants and wild-type zebrafish at three development stages (20, 45, and 60 dph) to perform transcriptomic analysis. The results showed that the deficiency of bmp6 upregulated sik1 and activated the TNF-A signaling via the NF-KB pathway, which inhibited the development of osteoblasts and promoted osteoclast formation, thereby inhibiting the formation of IBs. These results provided insights to understand the role of bmp6 in the development of IBs in zebrafish and are useful for selective breeding of IBs in cyprinids.
Collapse
Affiliation(s)
- Huan Xu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangxiang Tong
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| | - Ting Yan
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| | - Le Dong
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Xiaoxing Yang
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Dongyu Dou
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
| | - Zhipeng Sun
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| | - Tianqi Liu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| | - Xianhu Zheng
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| | - Jian Yang
- Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ningbo, China
| | - Xiaowen Sun
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| | - Yi Zhou
- Stem Cell Program of Boston Children’s Hospital, Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Youyi Kuang
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences, Harbin, China
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Harbin, China
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China
| |
Collapse
|
11
|
Song MK, Jung S, Hong S, Kwon JO, Kim MK, Kim HH. Effects of the Lysine Methyltransferase Inhibitor AZ505 on Bone Metabolism. J Bone Metab 2021; 28:297-305. [PMID: 34905676 PMCID: PMC8671023 DOI: 10.11005/jbm.2021.28.4.297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/13/2021] [Indexed: 11/25/2022] Open
Abstract
Background Protein methylation has important role in regulating diverse cellular responses, including differentiation, by affecting protein activity, stability, and interactions. AZ505 is an inhibitor of the SET and MYND domain-containing protein 2 lysine methylase. In this study, we investigated the effect of AZ505 on osteoblast and osteoclast differentiation in vitro and evaluated the effect of AZ505 in vivo on the long bones in mice. Methods Osteoblast differentiation was assessed by alkaline phosphatase (ALP) and Alizarin red staining after culturing calvarial preosteoblasts in an osteogenic medium. Osteoclast differentiation was analyzed by tartrate-resistant acid phosphatase (TRAP) staining in bone marrow-derived macrophages cultured with macrophage-colony stimulating factor and receptor activator of nuclear factor-κB ligand (RANKL). For in vivo experiments, mice were intraperitoneally injected with AZ505 and femurs were examined by micro-computed tomography. Results AZ505 increased ALP and Alizarin red staining in cultured osteoblasts and the expression of osteoblast marker genes, including Runx2 and osteocalcin. AZ505 resulted in decreased TRAP-staining of osteoclasts and expression of c-Fos and nuclear factor of activated T cells transcription factors and osteoclast marker genes, including cathepsin K and dendritic cell-specific transmembrane protein. Unexpectedly, in vivo administration of AZ505 markedly decreased the trabecular bone mass of femurs. In support of this catabolic result, AZ505 strongly upregulated RANKL expression in osteoblasts. Conclusions The results indicate that AZ505 has a catabolic effect on bone metabolism in vivo despite its anabolic effect in bone cell cultures. The findings indicate that cell culture data should be extrapolated cautiously to in vivo outcomes for studying bone metabolism.
Collapse
Affiliation(s)
- Min-Kyoung Song
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Suhan Jung
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Seojin Hong
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Jun-Oh Kwon
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| |
Collapse
|
12
|
Cohen P, Cross D, Jänne PA. Kinase drug discovery 20 years after imatinib: progress and future directions. Nat Rev Drug Discov 2021; 20:551-569. [PMID: 34002056 PMCID: PMC8127496 DOI: 10.1038/s41573-021-00195-4] [Citation(s) in RCA: 548] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 02/04/2023]
Abstract
Protein kinases regulate nearly all aspects of cell life, and alterations in their expression, or mutations in their genes, cause cancer and other diseases. Here, we review the remarkable progress made over the past 20 years in improving the potency and specificity of small-molecule inhibitors of protein and lipid kinases, resulting in the approval of more than 70 new drugs since imatinib was approved in 2001. These compounds have had a significant impact on the way in which we now treat cancers and non-cancerous conditions. We discuss how the challenge of drug resistance to kinase inhibitors is being met and the future of kinase drug discovery.
Collapse
Affiliation(s)
- Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK.
| | | | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana Farber Cancer Institute, Harvard University, Boston, MA, USA.
| |
Collapse
|
13
|
Sato T, Verma S, Khatri A, Dean T, Goransson O, Gardella TJ, Wein MN. Comparable Initial Engagement of Intracellular Signaling Pathways by Parathyroid Hormone Receptor Ligands Teriparatide, Abaloparatide, and Long-Acting PTH. JBMR Plus 2021; 5:e10441. [PMID: 33977197 PMCID: PMC8101618 DOI: 10.1002/jbm4.10441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple analogs of parathyroid hormone, all of which bind to the PTH/PTHrP receptor PTH1R, are used for patients with osteoporosis and hypoparathyroidism. Although ligands such as abaloparatide, teriparatide (hPTH 1-34 [TPTD]), and long-acting PTH (LA-PTH) show distinct biologic effects with respect to skeletal and mineral metabolism endpoints, the mechanistic basis for these clinically-important differences remains incompletely understood. Previous work has revealed that differential signaling kinetics and receptor conformation engagement between different PTH1R peptide ligands. However, whether such acute membrane proximal differences translate into differences in downstream signaling output remains to be determined. Here, we directly compared short-term effects of hPTH (1-34), abaloparatide, and LA-PTH in multiple cell-based PTH1R signaling assays. At the time points and ligand concentrations utilized, no significant differences were observed between these three ligands at the level of receptor internalization, β-arrestin recruitment, intracellular calcium stimulation, and cAMP generation. However, abaloparatide showed significantly quicker PTH1R recycling in washout studies. Downstream of PTH1R-stimulated cAMP generation, protein kinase A regulates gene expression via effects on salt inducible kinases (SIKs) and their substrates. Consistent with no differences between these ligands on cAMP generation, we observed that hPTH (1-34), abaloparatide, and LA-PTH showed comparable effects on SIK2 phosphorylation, SIK substrate dephosphorylation, and downstream gene expression changes. Taken together, these results indicate that these PTH1R peptide agonists engage downstream intracellular signaling pathways to a comparable degree. It is possible that differences observed in vivo in preclinical and clinical models may be related to pharmacokinetic factors. It is also possible that our current in vitro systems are insufficient to perfectly match the complexities of PTH1R signaling in bona fide target cells in bone in vivo. © 2020 American Society for Bone and Mineral Research © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Tadatoshi Sato
- Endocrine Unit, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Shiv Verma
- Endocrine Unit, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Ashok Khatri
- Endocrine Unit, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Thomas Dean
- Endocrine Unit, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Olga Goransson
- Department of Experimental Medical ScienceLund University, Diabetes, Metabolism and EndocrinologyLundSweden
| | - Thomas J Gardella
- Endocrine Unit, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Marc N Wein
- Endocrine Unit, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
14
|
Liu C, Han Y, Zhao X, Li B, Xu L, Li D, Li G. POLR2A blocks osteoclastic bone resorption and protects against osteoporosis by interacting with CREB1. J Cell Physiol 2021; 236:5134-5146. [PMID: 33595106 DOI: 10.1002/jcp.30220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 11/10/2022]
Abstract
Bone-resorbing osteoclasts significantly contribute to osteoporosis, and understanding the mechanisms of osteoclastogenesis is crucial for developing new drugs to treat diseases associated with bone loss. Here, we report that POLR2A is upregulated during osteoclastogenesis. Functional analyses showed that the inhibition of POLR2A decreased osteoclastogenesis, whereas the overexpression of POLR2A had completely opposite effects in vitro. Notably, the osteoclast-specific deletion of POLR2A blocks bone resorption in vivo. Furthermore, POLR2A loss-of-function suppresses estrogen deficiency-induced bone resorption. Mechanistically, POLR2A regulates the assembly of CREB1 on the regulatory elements of its target genes. Collectively, using genetic, pharmacological, and disease mouse models, we have identified a previously undescribed protein that interacts with CREB1 to regulate osteoclastic bone resorption.
Collapse
Affiliation(s)
- Chuxiao Liu
- State Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, People's Republic of China
| | - Yu Han
- Department of Joint Surgery, No. 1 Hospital of Jilin University, Changchun, People's Republic of China
| | - Xingyu Zhao
- Department of Joint Surgery, No. 1 Hospital of Jilin University, Changchun, People's Republic of China
| | - Bo Li
- State Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, People's Republic of China.,Department of Joint Surgery, No. 1 Hospital of Jilin University, Changchun, People's Republic of China
| | - Liwen Xu
- State Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, People's Republic of China
| | - Dongsong Li
- Department of Joint Surgery, No. 1 Hospital of Jilin University, Changchun, People's Republic of China
| | - Guangyu Li
- State Key Laboratory for Molecular Biology of Special Economic Animals, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, People's Republic of China
| |
Collapse
|
15
|
Tiyasatkulkovit W, Aksornthong S, Adulyaritthikul P, Upanan P, Wongdee K, Aeimlapa R, Teerapornpuntakit J, Rojviriya C, Panupinthu N, Charoenphandhu N. Excessive salt consumption causes systemic calcium mishandling and worsens microarchitecture and strength of long bones in rats. Sci Rep 2021; 11:1850. [PMID: 33473159 PMCID: PMC7817681 DOI: 10.1038/s41598-021-81413-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Excessive salt intake has been associated with the development of non-communicable diseases, including hypertension with several cardiovascular consequences. Although the detrimental effects of high salt on the skeleton have been reported, longitudinal assessment of calcium balance together with changes in bone microarchitecture and strength under salt loading has not been fully demonstrated. To address these unanswered issues, male Sprague-Dawley rats were fed normal salt diet (NSD; 0.8% NaCl) or high salt diet (HSD; 8% NaCl) for 5 months. Elevation of blood pressure, cardiac hypertrophy and glomerular deterioration were observed in HSD, thus validating the model. The balance studies were performed to monitor calcium input and output upon HSD challenge. The HSD-induced increase in calcium losses in urine and feces together with reduced fractional calcium absorption led to a decrease in calcium retention. With these calcium imbalances, we therefore examined microstructural changes of long bones of the hind limbs. Using the synchrotron radiation x-ray tomographic microscopy, we showed that trabecular structure of tibia and femur of HSD displayed a marked increase in porosity. Consistently, the volumetric micro-computed tomography also demonstrated a significant decrease in trabecular bone mineral density with expansion of endosteal perimeter in the tibia. Interestingly, bone histomorphometric analyses indicated that salt loading caused an increase in osteoclast number together with decreases in osteoblast number and osteoid volume. This uncoupling process of bone remodeling in HSD might underlie an accelerated bone loss and bone structural changes. In conclusion, long-term excessive salt consumption leads to impairment of skeletal mass and integrity possibly through negative calcium balance.
Collapse
Affiliation(s)
- Wacharaporn Tiyasatkulkovit
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.7922.e0000 0001 0244 7875Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Sirion Aksornthong
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Punyanuch Adulyaritthikul
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Pornpailin Upanan
- grid.411825.b0000 0000 9482 780XFaculty of Allied Health Sciences, Burapha University, Chonburi, 20131 Thailand
| | - Kannikar Wongdee
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.411825.b0000 0000 9482 780XFaculty of Allied Health Sciences, Burapha University, Chonburi, 20131 Thailand
| | - Ratchaneevan Aeimlapa
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Jarinthorn Teerapornpuntakit
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.412029.c0000 0000 9211 2704Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000 Thailand
| | - Catleya Rojviriya
- grid.472685.aSynchrotron Light Research Institute (Public Organization), Nakhon Ratchasima, 30000 Thailand
| | - Nattapon Panupinthu
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Narattaphol Charoenphandhu
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170 Thailand ,The Academy of Science, The Royal Society of Thailand, Dusit, Bangkok, 10300 Thailand
| |
Collapse
|
16
|
Sun Z, Niu S, Xu F, Zhao W, Ma R, Chen M. CircAMOTL1 Promotes Tumorigenesis Through miR-526b/SIK2 Axis in Cervical Cancer. Front Cell Dev Biol 2020; 8:568190. [PMID: 33344445 PMCID: PMC7744824 DOI: 10.3389/fcell.2020.568190] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/09/2020] [Indexed: 12/23/2022] Open
Abstract
Background Cervical cancer is one of the most common malignancies in women, leading to major health problems for its high morbidity and mortality. Numerous studies have demonstrated that circular RNAs (circRNAs) could be participated in the progression of multifarious diseases, especially plentiful carcinomas. CircAMOTL1 (angiomotin-like1, ID: hsa_circ_0004214), which is located on human chromosome 11:9 4532555-94533477, is involved in the occurrence of breast cancer, etc. However, the intrinsic and concrete molecular mechanism of circAMOTL1 in cervical carcinomas remained thoroughly unclear, which was also the bottleneck of circRNAs studies in cancer. Methods The relative expression levels of circAMOTL1 and miR-526b in cervical carcinoma patients’ specimens and cervical carcinoma cell lines were detected by RT-qPCR. Through experiments including loss-function and overexpression, the biological effects of circAMOTL1 and miR-526b on the proliferation, migration, apoptosis, and tumorigenicity were explored in cervical carcinomas. Dual luciferase reporter gene analysis, western blot, and other methods were adopted to explore the circAMOTL1 potential mechanism in cervical carcinomas. Results In our experiments, our researches displayed that circAMOTL1 was significantly higher expression in cervical carcinomas specimens and cell lines. Further experiments illustrated that the knockdown of circAMOTL1 could restrain the malignant phenotype, AKT signaling, and epithelial–mesenchymal transition (EMT) of in cervical carcinomas cells. Meanwhile miR-526b was downregulated in cervical carcinomas and even miR-526b could partially reverse circAMOTL1 function in malignant cervical tumor cells. CircAMOTL1 acts as a microRNA (miRNA) sponge that actively regulates the expression of salt-inducible kinase 2 (SIK2) to sponge miR-526b and subsequently increases malignant phenotypes of cervical carcinomas cells. In a word, circAMOTL1 acts a carcinogenic role and miR-526b serves as the opposite function of antioncogene in the cervical carcinoma pathogenesis. Conclusion CircAMOTL1-miR-526b-SIK2 axis referred to the malignant progression and development of cervical carcinomas. CircAMOTL1 expression was inversely correlated with miR-526b and positively correlated with SIK2 mRNA in cervical cancer tissues. Thus, circAMOTL1 exerted an oncogenic role in cervical cancer progression through sponging miR-526b. Taken together, our study revealed that circAMOTL1 acted as an oncogene and probably was a potential therapeutic target for the cervical cancer.
Collapse
Affiliation(s)
- Zhengwei Sun
- Department of Obstetrics & Gynecology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Sanqiang Niu
- Department of Obstetrics & Gynecology, Bozhou People's Hospital, Bozhou, China
| | - Fuxia Xu
- Department of Obstetrics & Gynecology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Weidong Zhao
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of USTC, Hefei, China
| | - Rong Ma
- Department of Obstetrics & Gynecology, Anhui Women and Child Health Care Hospital, Hefei, China
| | - Mingwei Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Sun Z, Jiang Q, Li J, Guo J. The potent roles of salt-inducible kinases (SIKs) in metabolic homeostasis and tumorigenesis. Signal Transduct Target Ther 2020; 5:150. [PMID: 32788639 PMCID: PMC7423983 DOI: 10.1038/s41392-020-00265-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/22/2020] [Indexed: 01/26/2023] Open
Abstract
Salt-inducible kinases (SIKs) belong to AMP-activated protein kinase (AMPK) family, and functions mainly involve in regulating energy response-related physiological processes, such as gluconeogenesis and lipid metabolism. However, compared with another well-established energy-response kinase AMPK, SIK roles in human diseases, especially in diabetes and tumorigenesis, are rarely investigated. Recently, the pilot roles of SIKs in tumorigenesis have begun to attract more attention due to the finding that the tumor suppressor role of LKB1 in non-small-cell lung cancers (NSCLCs) is unexpectedly mediated by the SIK but not AMPK kinases. Thus, here we tend to comprehensively summarize the emerging upstream regulators, downstream substrates, mouse models, clinical relevance, and candidate inhibitors for SIKs, and shed light on SIKs as the potential therapeutic targets for cancer therapies.
Collapse
Affiliation(s)
- Zicheng Sun
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China.,Department of Breast and Thyroid Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Qiwei Jiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China.
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China.
| |
Collapse
|
18
|
Xiong Y, Chen L, Yan C, Zhou W, Yu T, Sun Y, Cao F, Xue H, Hu Y, Chen D, Mi B, Liu G. M2 Macrophagy-derived exosomal miRNA-5106 induces bone mesenchymal stem cells towards osteoblastic fate by targeting salt-inducible kinase 2 and 3. J Nanobiotechnology 2020; 18:66. [PMID: 32345321 PMCID: PMC7189726 DOI: 10.1186/s12951-020-00622-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/21/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Osteoblast differentiation is a vital process for fracture healing, and exosomes are nanosized membrane vesicles that can deliver therapeutic drugs easily and safely. Macrophages participate in the regulation of various biological processes in vivo, and macrophage-derived exosomes (MD-Exos) have recently been a topic of increasing research interest. However, few study has explored the link between MD-Exos and osteoblast differentiation. Herein, we sought to identify miRNAs differentially expressed between M1 and M2 macrophage-derived exosomes, and to evaluate their roles in the context of osteoblast differentiation. RESULTS We found that microRNA-5106 (miR-5106) was significantly overexpressed in M2 macrophage-derived exosomes (M2D-Exos), while its expression was decreased in M1 macrophage-derived exosomes (M1D-Exos), and we found that this exosomal miRNA can induce bone mesenchymal stem cell (BMSC) osteogenic differentiation via directly targeting the Salt-inducible kinase 2 and 3 (SIK2 and SIK3) genes. In addition, the local injection of both a miR-5106 agonist or M2D-Exos to fracture sites was sufficient to accelerate healing in vivo. CONCLUSIONS Our study demonstrates that miR-5106 is highly enriched in M2D-Exos, and that it can be transferred to BMSCs wherein it targets SIK2 and SIK3 genes to promote osteoblast differentiation.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chenchen Yan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Yu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yun Sun
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Faqi Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hang Xue
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiqiang Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dong Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|