1
|
Wang X, Qu Z, Zhao S, Luo L, Yan L. Wnt/β-catenin signaling pathway: proteins' roles in osteoporosis and cancer diseases and the regulatory effects of natural compounds on osteoporosis. Mol Med 2024; 30:193. [PMID: 39468464 PMCID: PMC11520425 DOI: 10.1186/s10020-024-00957-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: 05/17/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Osteoblasts are mainly derived from mesenchymal stem cells in the bone marrow. These stem cells can differentiate into osteoblasts, which have the functions of secreting bone matrix, promoting bone formation, and participating in bone remodeling. The abnormality of osteoblasts can cause a variety of bone-related diseases, including osteoporosis, delayed fracture healing, and skeletal deformities. In recent years, with the side effects caused by the application of PTH drugs, biphosphonate drugs, and calmodulin drugs, people have carried out more in-depth research on the mechanism of osteoblast differentiation, and are actively looking for natural compounds for the treatment of osteoporosis. The Wnt/β-catenin signaling pathway is considered to be one of the important pathways of osteoblast differentiation, and has become an important target for the treatment of osteoporosis. The Wnt/β-catenin signaling pathway, whether its activation is enhanced or its expression is weakened, will cause a variety of diseases including tumors. This review will summarize the effect of Wnt/β-catenin signaling pathway on osteoblast differentiation and the correlation between the related proteins in the pathway and human diseases. At the same time, the latest research progress of natural compounds targeting Wnt/β-catenin signaling pathway against osteoporosis is summarized.
Collapse
Affiliation(s)
- Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Medical University, Xi'an, China
| | - Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Luo
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Ma Y, Su H, Li W, Mao S, Feng Z, Qiu Y, Chen K, Chen Q, Wang H, Zhu Z. The hyaluronic acid-gelatin hierarchical hydrogel for osteoporotic bone defect repairment. Int J Biol Macromol 2024; 276:133821. [PMID: 38996892 DOI: 10.1016/j.ijbiomac.2024.133821] [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: 03/29/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Osteoporotic bone defects are serious medical problems due to their sparse bone structure, difficulty in restoration and reconstruction, and high recurrence rates, which also result in heavy economic and social burdens. Herein, we developed a hierarchical hydrogel composed of alendronate sodium (AS)/Mg2+-loaded inverse opal methylpropenylated gelatin (GelMA) hydrogel microspheres (IOHM-AS-Mgs) within methylpropenylated poly(hyaluronic acid) (HAMA) for osteoporotic bone defect treatment. The IOHM-AS-Mgs displayed good cytocompatibility and cell adhesion and strongly stimulated osteogenesis at the transcriptomic and protein levels. When this treatment was applied to the osteoporotic bone defect area, HAMA was used to fix the microspheres. The results of the microcomputed tomography (micro-CT) and histological analyses indicated that the hierarchical hydrogel had the best therapeutic effect. Therefore, this hydrogel is a new candidate for osteoporotic bone defect treatment.
Collapse
Affiliation(s)
- Yanyu Ma
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Haiwen Su
- Department of Nephrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524013, China
| | - Wenhan Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Saihu Mao
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Zhenghua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Keng Chen
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| | - Quanchi Chen
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Huan Wang
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China.
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China.
| |
Collapse
|
3
|
Kitajima Y, Yoshioka K, Mikumo Y, Ohki S, Maehara K, Ohkawa Y, Ono Y. Loss of Tob1 promotes muscle regeneration through muscle stem cell expansion. J Cell Sci 2024; 137:jcs261886. [PMID: 39037211 DOI: 10.1242/jcs.261886] [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: 12/11/2023] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Muscle stem cells (MuSCs) play an indispensable role in postnatal muscle growth and hypertrophy in adults. MuSCs also retain a highly regenerative capacity and are therefore considered a promising stem cell source for regenerative therapy for muscle diseases. In this study, we identify tumor-suppressor protein Tob1 as a Pax7 target protein that negatively controls the population expansion of MuSCs. Tob1 protein is undetectable in the quiescent state but is upregulated during activation in MuSCs. Tob1 ablation in mice accelerates MuSC population expansion and boosts muscle regeneration. Moreover, inactivation of Tob1 in MuSCs ameliorates the efficiency of MuSC transplantation in a murine muscular dystrophy model. Collectively, selective targeting of Tob1 might be a therapeutic option for the treatment of muscular diseases, including muscular dystrophy and age-related sarcopenia.
Collapse
Affiliation(s)
- Yasuo Kitajima
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Yoko Mikumo
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
| | - Shun Ohki
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Kazumitsu Maehara
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, 860-0811, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Division of Biological Regulation, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), Tokyo, 173-0015, Japan
| |
Collapse
|
4
|
Chen J, Liu C, Yang Y, Gong X, Qian H. The stratum corneum barrier: impaired function in relation to associated lipids and proteins. Tissue Barriers 2024:2361197. [PMID: 38818698 DOI: 10.1080/21688370.2024.2361197] [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: 04/03/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024] Open
Abstract
The skin is the largest organ of the human body and is widely considered to be the first-line defense of the body, providing essential protection against mechanical, physical, and chemical damage. Keratinocytes are the primary cells of the outer layer of the epidermis, which acts as a mechanical and permeability barrier. The epidermis is a permanently renewed tissue where undifferentiated keratinocytes located at the basal layer proliferate and migrate to the overlying layers. Here we report that some components of keratinocytes affect the formation and differentiation of the stratum corneum, which is the most specialized layer of the epidermis.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, Xuhui, District, China
| | - Changjie Liu
- SIMPLY THIS Skin Ecology Research Institute, Shili (Shanghai) Biotechology Co., Ltd, Shanghai, China
| | - Yuan Yang
- SIMPLY THIS Skin Ecology Research Institute, Shili (Shanghai) Biotechology Co., Ltd, Shanghai, China
| | - Xue Gong
- SIMPLY THIS Skin Ecology Research Institute, Shili (Shanghai) Biotechology Co., Ltd, Shanghai, China
| | - Huan Qian
- Department of Plastic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Wu M, Wu S, Chen W, Li YP. The roles and regulatory mechanisms of TGF-β and BMP signaling in bone and cartilage development, homeostasis and disease. Cell Res 2024; 34:101-123. [PMID: 38267638 PMCID: PMC10837209 DOI: 10.1038/s41422-023-00918-9] [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: 02/26/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - 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
| | - 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
|
6
|
Perez RC, Yang X, Familari M, Martinez G, Lovicu FJ, Hime GR, de Iongh RU. TOB1 and TOB2 mark distinct RNA processing granules in differentiating lens fiber cells. J Mol Histol 2024; 55:121-138. [PMID: 38165569 DOI: 10.1007/s10735-023-10177-y] [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: 07/18/2023] [Accepted: 11/12/2023] [Indexed: 01/04/2024]
Abstract
Differentiation of lens fiber cells involves a complex interplay of signals from growth factors together with tightly regulated gene expression via transcriptional and post-transcriptional regulators. Various studies have demonstrated that RNA-binding proteins, functioning in ribonucleoprotein granules, have important roles in regulating post-transcriptional expression during lens development. In this study, we examined the expression and localization of two members of the BTG/TOB family of RNA-binding proteins, TOB1 and TOB2, in the developing lens and examined the phenotype of mice that lack Tob1. By RT-PCR, both Tob1 and Tob2 mRNA were detected in epithelial and fiber cells of embryonic and postnatal murine lenses. In situ hybridization showed Tob1 and Tob2 mRNA were most intensely expressed in the early differentiating fibers, with weaker expression in anterior epithelial cells, and both appeared to be downregulated in the germinative zone of E15.5 lenses. TOB1 protein was detected from E11.5 to E16.5 and was predominantly detected in large cytoplasmic puncta in early differentiating fiber cells, often co-localizing with the P-body marker, DCP2. Occasional nuclear puncta were also observed. By contrast, TOB2 was detected in a series of interconnected peri-nuclear granules, in later differentiating fiber cells of the inner cortex. TOB2 did not appear to co-localize with DCP2 but did partially co-localize with an early stress granule marker (EIF3B). These data suggest that TOB1 and TOB2 are involved with different aspects of the mRNA processing cycle in lens fiber cells. In vitro experiments using rat lens epithelial explants treated with or without a fiber differentiating dose of FGF2 showed that both TOB1 and TOB2 were up-regulated during FGF-induced differentiation. In differentiating explants, TOB1 also co-localized with DCP2 in large cytoplasmic granules. Analyses of Tob1-/- mice revealed relatively normal lens morphology but a subtle defect in cell cycle arrest of some cells at the equator and in the lens fiber mass of E13.5 embryos. Overall, these findings suggest that TOB proteins play distinct regulatory roles in RNA processing during lens fiber differentiation.
Collapse
Affiliation(s)
- Rafaela C Perez
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Xenia Yang
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Mary Familari
- School of Biosciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Gemma Martinez
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Frank J Lovicu
- Molecular and Cellular Biomedicine, School of Medical Sciences and Save Sight Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Gary R Hime
- Stem Cell Genetics Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Robb U de Iongh
- Ocular Development Laboratory, Anatomy & Physiology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
7
|
Xie Z, Zhou G, Zhang M, Han J, Wang Y, Li X, Wu Q, Li M, Zhang S. Recent developments on BMPs and their antagonists in inflammatory bowel diseases. Cell Death Discov 2023; 9:210. [PMID: 37391444 DOI: 10.1038/s41420-023-01520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis, and Crohn's disease, are intestinal disorders characterized by chronic relapsing inflammation. A large proportion of patients with IBD will progress to develop colitis-associated colorectal cancer due to the chronic intestinal inflammation. Biologic agents that target tumour necrosis factor-α, integrin α4β7, and interleukin (IL)12/23p40 have been more successful than conventional therapies in treating IBD. However, drug intolerance and loss of response are serious drawbacks of current biologics, necessitating the development of novel drugs that target specific pathways in IBD pathogenesis. One promising group of candidate molecules are bone morphogenetic proteins (BMPs), members of the TGF-β family involved in regulating morphogenesis, homeostasis, stemness, and inflammatory responses in the gastrointestinal tract. Also worth examining are BMP antagonists, major regulators of these proteins. Evidence has shown that BMPs (especially BMP4/6/7) and BMP antagonists (especially Gremlin1 and follistatin-like protein 1) play essential roles in IBD pathogenesis. In this review, we provide an updated overview on the involvement of BMPs and BMP antagonists in IBD pathogenesis and in regulating the fate of intestinal stem cells. We also described the expression patterns of BMPs and BMP antagonists along the intestinal crypt-villus axis. Lastly, we synthesized available research on negative regulators of BMP signalling. This review summarizes recent developments on BMPs and BMP antagonists in IBD pathogenesis, which provides novel insights into future therapeutic strategies.
Collapse
Affiliation(s)
- Zhuo Xie
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Gaoshi Zhou
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Mudan Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jing Han
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Ying Wang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiaoling Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Qirui Wu
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Manying Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China.
| |
Collapse
|
8
|
Lu L, Sun S, Li H, Xie Y. Functional mechanism of miR-92b-3p in osteogenic differentiation of fibroblasts in patients with ankylosing spondylitis via the TOB1/BMP/Smad pathway. J Orthop Surg Res 2023; 18:402. [PMID: 37268992 DOI: 10.1186/s13018-023-03850-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 05/10/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic inflammatory arthritis. Upregulation of microRNA (miR)-92b-3p is associated with enhanced osteoblastic differentiation. The current study sought to investigate the functional mechanism of miR-92b-3p in osteogenic differentiation of AS fibroblasts. METHODS First, fibroblasts were isolated from AS and non-AS patients and cultured. Next, cell morphology was observed, cell proliferation was assessed and the vimentin expression pattern was determined. Alkaline phosphatase (ALP) activity and levels of osteogenic markers RUNX2, OPN, OSX, and COL I were additionally measured, followed by determination of miR-92b-3p and TOB1 levels. The binding site of miR-92b-3p and TOB1 was predicted, and their target relationship was validated. Lastly, miR-92b-3p inhibitor, si-TOB1, and the BMP/Smad signaling pathway inhibitor LDN193189 were delivered into AS fibroblasts to evaluate the osteogenic differentiation of AS fibroblasts and the activation of the BMP/Smad pathway. RESULTS miR-92b-3p was highly expressed in AS fibroblasts. AS fibroblasts showed enhanced osteogenic differentiation and proliferation, while inhibition of miR-92b-3p suppressed osteogenic differentiation and proliferation of AS fibroblasts. miR-92b-3p targeted TOB1, and TOB1 was poorly expressed in AS fibroblasts. The concurrent downregulation of TOB1 and inhibition of miR-92b-3p elevated the levels of RUNX2, OPN, OSX, and COL I and ALP activity and further enhanced the proliferation of AS fibroblasts. The BMP/Smad pathway was activated in AS fibroblasts. Silencing miR-92b-3p could inhibit the activation of the BMP/Smad pathway by upregulating TOB1. Inhibition of the BMP/Smad pathway reduced the number of calcified nodules and hindered the osteogenic differentiation and proliferation of AS fibroblasts. CONCLUSION Our findings highlighted that silencing miR-92b-3p inhibited the osteogenic differentiation and proliferation of AS fibroblasts by upregulation of TOB1 and inhibition of the BMP/Smad pathway.
Collapse
Affiliation(s)
- Liansong Lu
- Department of Spinal Surgery, Ningbo No.6 Hospital, 1059 East Zhongshan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China.
| | - Shaohua Sun
- Department of Spinal Surgery, Ningbo No.6 Hospital, 1059 East Zhongshan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Haoji Li
- Department of Spinal Surgery, Ningbo No.6 Hospital, 1059 East Zhongshan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| | - Yingzhi Xie
- Department of Medical Image, Ningbo No.6 Hospital, 1059 East Zhongshan Road, Yinzhou District, Ningbo, 315040, Zhejiang, China
| |
Collapse
|
9
|
Nucleus pulposus related lncRNA and mRNA expression profiles in intervertebral disc degeneration. Genomics 2023; 115:110570. [PMID: 36746221 DOI: 10.1016/j.ygeno.2023.110570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023]
Abstract
In the present study, we aimed to have a comprehensive understanding of nucleus pulposus related long noncoding RNA (lncRNA) and mRNA expression profiles in intervertebral disc degeneration (IDD). In total, 2418 mRNAs and 528 lncRNAs were found to be differentially expressed in the IDD group compared with the Control group. Combining microarray datasets and sequencing data, 5 overlapping DEMs and 7 overlapping DELs were identified. NF-κB signaling pathway, PI3K-Akt signaling pathway and Wnt/β-catenin signaling pathway were strongly linked with enriched GO terms and KEGG pathways. The ceRNA network suggested that lnc-TMEM44-AS1-hsa-miR-206-HDAC4 may be one crucial axis in IDD. PPI network analysis was constructed with 309 nodes and 129 edges. And the highest connectivity degrees were ALB, APOB and CCL2. This study suggested that specific lncRNAs and ceRNA axes may be crucial in the development of IDD. It provides a new perspective for delaying IDD process and enhancing intervertebral disc repair.
Collapse
|
10
|
Yu Z, Wang L, Zhao J, Song H, Zhao C, Zhao W, Jia M. TOB1 attenuates IRF3-directed antiviral responses by recruiting HDAC8 to specifically suppress IFN-β expression. Commun Biol 2022; 5:943. [PMID: 36085336 PMCID: PMC9463440 DOI: 10.1038/s42003-022-03911-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Interferon regulatory factor 3 (IRF3) is a key transcription factor required for the secretion of type I interferons (IFN-α/β) and initiation of antiviral immune response. However, the negative feedback regulator of IRF3-directed antiviral response remains unknown. In this study, we demonstrated that viral infection induced the interaction of the transducer of ERBB2.1 (TOB1) with IRF3, which bound to the promoter region of Ifnb1 in macrophages. TOB1 inhibited Ifnb1 transcription by disrupting IRF3 binding and recruiting histone deacetylase 8 (HDAC8) to the Ifnb1 promoter region. Consequently, TOB1 attenuated IRF3-directed IFN-β expression in virus-infected macrophages. Tob1 deficiency enhanced antiviral response and suppressed viral replication in vivo. Thus, we identified TOB1 as a feedback inhibitor of host antiviral innate immune response and revealed a mechanism underlying viral immune escape. TOB1 is identified as an interferon regulatory factor 3 (IRF3) binding partner that operates as a negative feedback inhibitor of IFNβ in toll-like receptor and cytosolic nucleic acid receptor sensing pathways.
Collapse
|
11
|
Youssef MMM, Hamada HT, Lai ESK, Kiyama Y, El-Tabbal M, Kiyonari H, Nakano K, Kuhn B, Yamamoto T. TOB is an effector of the hippocampus-mediated acute stress response. Transl Psychiatry 2022; 12:302. [PMID: 35906220 PMCID: PMC9338090 DOI: 10.1038/s41398-022-02078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Stress affects behavior and involves critical dynamic changes at multiple levels ranging from molecular pathways to neural circuits and behavior. Abnormalities at any of these levels lead to decreased stress resilience and pathological behavior. However, temporal modulation of molecular pathways underlying stress response remains poorly understood. Transducer of ErbB2.1, known as TOB, is involved in different physiological functions, including cellular stress and immediate response to stimulation. In this study, we investigated the role of TOB in psychological stress machinery at molecular, neural circuit, and behavioral levels. Interestingly, TOB protein levels increased after mice were exposed to acute stress. At the neural circuit level, functional magnetic resonance imaging (fMRI) suggested that intra-hippocampal and hippocampal-prefrontal connectivity were dysregulated in Tob knockout (Tob-KO) mice. Electrophysiological recordings in hippocampal slices showed increased postsynaptic AMPAR-mediated neurotransmission, accompanied by decreased GABA neurotransmission and subsequently altered Excitatory/Inhibitory balance after Tob deletion. At the behavioral level, Tob-KO mice show abnormal, hippocampus-dependent, contextual fear conditioning and extinction, and depression-like behaviors. On the other hand, increased anxiety observed in Tob-KO mice is hippocampus-independent. At the molecular level, we observed changes in factors involved in stress response like decreased stress-induced LCN2 expression and ERK phosphorylation, as well as increased MKP-1 expression. This study introduces TOB as an important modulator in the hippocampal stress signaling machinery. In summary, we reveal a molecular pathway and neural circuit mechanism by which Tob deletion contributes to expression of pathological stress-related behavior.
Collapse
Affiliation(s)
- Mohieldin M M Youssef
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| | - Hiro Taiyo Hamada
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Esther Suk King Lai
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yuji Kiyama
- Laboratory of Biochemistry and Molecular Biology, Graduate school of medical and dental sciences, Kagoshima University, Kagoshima, Japan
| | - Mohamed El-Tabbal
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Kohei Nakano
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Bernd Kuhn
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
12
|
Lin R, Ma C, Fang L, Xu C, Zhang C, Wu X, Wu W, Zhu R, Cong Y, Liu Z. TOB1 Blocks Intestinal Mucosal Inflammation Through Inducing ID2-Mediated Suppression of Th1/Th17 Cell Immune Responses in IBD. Cell Mol Gastroenterol Hepatol 2021; 13:1201-1221. [PMID: 34920145 PMCID: PMC8881672 DOI: 10.1016/j.jcmgh.2021.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS TOB1 is an anti-proliferative protein of Tob/BTG family and typically involved in the tumorigenesis and T cell activation. Although TOB1 is associated with T helper 17 cell-related autoimmunity, its role in modulating T cell-mediated immune responses in IBD remains poorly understood. Here, we explored its expression and the underlying mechanisms involved in the pathogenesis of inflammatory bowel disease (IBD). METHODS TOB1 and ID2 expression in IBD patients was examined by quantitative real time polymerase chain reaction and immunohistochemistry. IBD CD4+ T cells were transfected with lentivirus expressing TOB1, ID2, TOB1 short hairpin RNA and ID2 short hairpin RNA, respectively, and Tob1-/-CD4+ T cells were transfected with lentivirus expressing Id2. Experimental colitis was established in Tob1-/- mice by trinitrobenzene sulfonic acid enema and in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells to further explore the role of Tob1 in intestinal mucosal inflammation. Splenic CD4+ T cells of Tob1-/- mice were sorted to determine transcriptome differences by RNA sequencing. RESULTS TOB1 expression was decreased in inflamed mucosa and peripheral blood CD4+ T cells of IBD patients compared with healthy subjects. Overexpression of TOB1 downregulated IBD CD4+ T cells to differentiate into Th1/Th17 cells compared with control subjects. Severe colitis was observed in Tob1-/- mice through trinitrobenzene sulfonic acid enema or in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells, compared with control animals. RNA sequencing analysis revealed ID2 as functional target of TOB1 to inhibit IBD CD4+ T cell differentiation into Th1/Th17 cells. Mechanistically, TOB1 was associated with Smad4/5 to induce ID2 expression and restrain Th1/Th17 cell differentiation. CONCLUSIONS TOB1 restrains intestinal mucosal inflammation through suppressing Th1/Th17 cell-mediated immune responses via the Smad4/5-ID2 pathway. It may serve as a novel therapeutic target for treatment of human IBD.
Collapse
Affiliation(s)
- Ritian Lin
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Caiyun Ma
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Leilei Fang
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunjin Xu
- Department of Gastroenterology, First People’s Hospital of Shangqiu City Affiliated to Xinxiang Medical University, Shangqiu, China
| | - Cui Zhang
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaohan Wu
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Wu
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ruixin Zhu
- Department of Bioinformatics, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Zhanju Liu
- Center for IBD Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Correspondence Address correspondence to: Zhanju Liu, MD, PhD, Center for IBD Research, The Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China. fax: +86 21 66303983.
| |
Collapse
|
13
|
Overexpression Effects of miR-424 and BMP2 on the Osteogenesis of Wharton's Jelly-Derived Stem Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7031492. [PMID: 34790821 PMCID: PMC8592721 DOI: 10.1155/2021/7031492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023]
Abstract
Recently, the translational application of noncoding RNAs is accelerated dramatically. In this regard, discovering therapeutic roles of microRNAs by developing synthetic RNA and vector-based RNA is attracting attention. Here, we studied the effect of BMP2 and miR-424 on the osteogenesis of Wharton's jelly-derived stem cells (WJSCs). For this purpose, human BMP2 and miR-424 DNA codes were cloned in the third generation of lentiviral vectors and then used for HEK-293T cell transfection. Lentiviral plasmids contained miR424, BMP-2, miR424-BMP2, green fluorescent protein (GFP) genes, and helper vectors. The recombinant lentiviral particles transduced the WJSCs, and the osteogenesis was evaluated by real-time PCR, Western blot, Alizarin Red staining, and alkaline phosphatase enzyme activity. According to the results, there was a significant increase in the expression of the BMP2 gene and secretion of Osteocalcin protein in the group of miR424-BMP2. Moreover, the amount of dye deposition in Alizarin Red staining and alkaline phosphatase activity was significantly higher in the mentioned group (p < 0.05). Thus, the current study results clarify the efficacy of gene therapy by miR424-BMP2 vectors for bone tissue engineering. These data could help guide the development of gene therapy-based protocols for bone tissue engineering.
Collapse
|
14
|
Biradar VS, Rajpathak SN, Joshi SR, Deobagkar DD. Functional and regulatory aspects of oxidative stress response in X monosomy. In Vitro Cell Dev Biol Anim 2021; 57:661-675. [PMID: 34505228 DOI: 10.1007/s11626-021-00604-3] [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: 04/28/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022]
Abstract
The partial/complete loss of one X chromosome in a human female leads to Turner syndrome (TS). TS individuals display a range of phenotypes including short stature, osteoporosis, ovarian malfunction, diabetes, and thyroid dysfunction. Epigenetic factors and regulatory networks are distinctly different in X monosomy (45, X). In a lifetime, an individual is exposed to a variety of stress conditions. To study whether X monosomy cells display a differential response upon exposure to mild stress as compared to normal 46, XX cells and whether this may contribute to various co-morbidities in aneuploid individuals, we have carried out a transcriptomic analysis of human fibroblasts 45, X and 46, XX after exposure to mild oxidative stress. Under these conditions, over 350 transcripts were seen to be differentially expressed in 45, X and 46, XX cells. Pathways associated with oxidative stress were differentially expressed highlighting the differential regulation of genes and associated phenotypes. It could be seen that X monosomy cells are more susceptible to oxidative stress as compared to normal cells and have altered molecular pathways both in normal conditions and also upon exposure to mild oxidative stress. To explore this aspect in detail, we have mapped the expressions of transcription factors (TFs) in 45, X and 46, XX cells. The network of transcription activating factors is differentially regulated in 45, X and 46, XX cells under stress exposure. It is tempting to speculate that the altered ability of 45, X (Turner) cells to respond to stress may play a significant role in the physiological function and altered phenotypes in Turner syndrome.
Collapse
Affiliation(s)
- Vinayak S Biradar
- Molecular Biology Research Laboratory, Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Shriram N Rajpathak
- Molecular Biology Research Laboratory, Department of Zoology, Savitribai Phule Pune University, Pune, India
- Recombinant Department, Serum Institute of India Pvt. Ltd., Pune, 411 028, India
| | - Suraj R Joshi
- Molecular Biology Research Laboratory, Department of Zoology, Savitribai Phule Pune University, Pune, India
| | - Deepti D Deobagkar
- Molecular Biology Research Laboratory, Department of Zoology, Savitribai Phule Pune University, Pune, India.
- School of Physical Sciences, ISRO Space Technology Cell, Savitribai Phule Pune University, Pune, 411 007, India.
| |
Collapse
|
15
|
Cyclosporine A Promotes Bone Remodeling in LPS-Related Inflammation via Inhibiting ROS/ERK Signaling: Studies In Vivo and In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8836599. [PMID: 33505590 PMCID: PMC7810558 DOI: 10.1155/2021/8836599] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 02/05/2023]
Abstract
In some inflammatory diseases of bone, osteogenesis and osteoclasis are uncoupled and the balance is usually tipped resulting in bone destruction. The underlying mechanism of osteogenic dysfunction in inflammation still needs further study. This study is aimed at investigating the effects of cyclosporine A (CsA) on bone remodeling in lipopolysaccharide- (LPS-) related inflammation. In vivo, an alveolar bone defect model was established using 10-week-old C57BL/6J mice. The mice were divided into phosphate-buffered saline (PBS), LPS, and LPS+CsA groups. After 3 weeks, micro-CT analysis and histomorphometric evaluation were conducted. In vitro, murine osteoblasts were treated with vehicle medium, LPS, LPS+CsA, LPS+extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor (LPS+PD98059), and LPS+antioxidant (LPS+EUK134). Cell proliferation, osteogenic behaviors, oxidative stress, and ERK signaling were determined. By these approaches, LPS inhibited bone remodeling and promoted oxidative stress accumulation in alveolar bone defects. When animals were treated with CsA, all LPS-induced biochemical changes ameliorated with a marked protective effect. In vitro, the reactive oxygen species (ROS) levels in mitochondria increased in LPS-treated osteoblasts, with decreased expression of osteogenic differentiation genes. The CsA, PD98059, and EUK134 presented remarkable protective effects against LPS treatment. CsA effectively enhanced bone remodeling and attenuated oxidative stress caused by LPS via inhibiting ROS/ERK signaling. Taken together, the protective effect of CsA and the inhibitory effect of ERK signaling on the maintenance of mitochondrial function and reduction of ROS levels hold promise as a potential novel therapeutic strategy for inflammatory diseases in bones.
Collapse
|
16
|
Kanakamani S, Suresh PS, Venkatesh T. Regulation of processing bodies: From viruses to cancer epigenetic machinery. Cell Biol Int 2020; 45:708-719. [PMID: 33325125 DOI: 10.1002/cbin.11527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/17/2020] [Accepted: 12/13/2020] [Indexed: 11/08/2022]
Abstract
Processing bodies (PBs) are 100-300 nm cytoplasmic messenger ribonucleoprotein particle (mRNP) granules that regulate eukaryotic gene expression. These cytoplasmic compartments harbor messenger RNAs (mRNAs) and several proteins involved in mRNA decay, microRNA silencing, nonsense-mediated mRNA decay, and splicing. Though membrane-less, PB structures are maintained by RNA-protein and protein-protein interactions. PB proteins have intrinsically disordered regions and low complexity domains, which account for its liquid to liquid phase separation. In addition to being dynamic and actively involved in the exchange of materials with other mRNPs and organelles, they undergo changes on various cellular cues and environmental stresses, including viral infections. Interestingly, several PB proteins are individually implicated in cancer development, and no study has addressed the effects on PB dynamics after epigenetic modifications of cancer-associated PB genes. In the current review, we summarize modulations undergone by P bodies or P body components upon viral infections. Furthermore, we discuss the selective and widely investigated PB proteins that undergo methylation changes in cancer and their potential as biomarkers.
Collapse
Affiliation(s)
- Sunmathy Kanakamani
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, India
| | - Padmanaban S Suresh
- Department of Biotechnology, National Institute of Technology Calicut, Calicut, India
| | - Thejaswini Venkatesh
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, India
| |
Collapse
|
17
|
Ramaswamy Y, Roohani I, No YJ, Madafiglio G, Chang F, Zhang F, Lu Z, Zreiqat H. Nature-inspired topographies on hydroxyapatite surfaces regulate stem cells behaviour. Bioact Mater 2020; 6:1107-1117. [PMID: 33102949 PMCID: PMC7569262 DOI: 10.1016/j.bioactmat.2020.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
Surface topography is one of the key factors in regulating interactions between materials and cells. While topographies presented to cells in vivo are non-symmetrical and in complex shapes, current fabrication techniques are limited to replicate these complex geometries. In this study, we developed a microcasting technique and successfully produced imprinted hydroxyapatite (HAp) surfaces with nature-inspired (honeycomb, pillars, and isolated islands) topographies. The in vitro biological performance of the developed non-symmetrical topographies was evaluated using adipose-derived stem cells (ADSCs). We demonstrated that ADSCs cultured on all HAp surfaces, except honeycomb patterns, presented well-defined stress fibers and expressed focal adhesion protein (paxillin) molecules. Isolated islands topographies significantly promoted osteogenic differentiation of ADSCs with increased alkaline phosphatase activity and upregulation of key osteogenic markers, compared to the other topographies and the control unmodified (flat) HAp surface. In contrast, honeycomb topographies hampered the ability of the ADSCs to proliferate and differentiate to the osteogenic lineage. This work presents a facile technique to imprint nature-derived topographies on the surface of bioceramics which opens up opportunities for the development of bioresponsive interfaces in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Yogambha Ramaswamy
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia.,Australian Research Centre for Innovative BioEngineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Iman Roohani
- School of Chemistry, Australian Centre for Nanomedicine, University of New South Wales, Sydney NSW, Australia
| | - Young Jung No
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia.,Australian Research Centre for Innovative BioEngineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Genevieve Madafiglio
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Frank Chang
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Furong Zhang
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Zufu Lu
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia.,Australian Research Centre for Innovative BioEngineering, University of Sydney, Sydney, NSW, 2006, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of Biomedical Engineering, University of Sydney, Sydney, NSW, 2006, Australia.,Australian Research Centre for Innovative BioEngineering, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
18
|
Bai J, Wang H, Chen H, Ge G, Wang M, Gao A, Tong L, Xu Y, Yang H, Pan G, Chu PK, Geng D. Biomimetic osteogenic peptide with mussel adhesion and osteoimmunomodulatory functions to ameliorate interfacial osseointegration under chronic inflammation. Biomaterials 2020; 255:120197. [DOI: 10.1016/j.biomaterials.2020.120197] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/05/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023]
|
19
|
Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 2020; 21:696-711. [PMID: 32901139 DOI: 10.1038/s41580-020-00279-w] [Citation(s) in RCA: 473] [Impact Index Per Article: 118.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment - including inflammatory, endothelial and Schwann cells - persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.
Collapse
Affiliation(s)
- Ankit Salhotra
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Harsh N Shah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
20
|
Jiang G, Gong M, Song H, Sun W, Zhao W, Wang L. Tob2 Inhibits TLR-Induced Inflammatory Responses by Association with TRAF6 and MyD88. THE JOURNAL OF IMMUNOLOGY 2020; 205:981-986. [PMID: 32611726 DOI: 10.4049/jimmunol.2000057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/08/2020] [Indexed: 12/16/2022]
Abstract
Optimal activation of TLR pathways is crucial for the initiation of inflammatory responses and eliminating invading micro-organisms. However, excessive of TLR activation may lead to autoimmune and inflammatory diseases. Thus, TLR pathways should be tightly controlled. In this study, we identify Tob2, a Tob/BTG family member, as a suppressor of TLR pathways. Tob2 deficiency enhances TLR-induced NF-κB and MAPK activation and promotes the expression of proinflammatory cytokines in primary peritoneal macrophages of C57BL/6 mice. Furthermore, Tob2-defective C57BL/6 mice may be more susceptible to endotoxemic shock in vivo. Mechanistically, Tob2 interacts with TRAF6 and MyD88 and thus inhibits signaling from the MyD88-TRAF6 complex in primary peritoneal macrophages and HEK293T cells. Therefore, our results uncover a regulatory mechanism of TLR pathways and provide a potential target for the intervention of diseases with excessive TLR activation.
Collapse
Affiliation(s)
- Guosheng Jiang
- Department of Immunology, College of Basic Medical, Binzhou Medical University, Yantai 256600, Shandong, China;
| | - Mouchun Gong
- Department of General Surgery, Lin'an District People's Hospital, Hangzhou 310013, Zhejiang, China
| | - Hui Song
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan 250012, Shandong, China; and
| | - Wangnan Sun
- Department of Immunology, College of Basic Medical, Binzhou Medical University, Yantai 256600, Shandong, China
| | - Wei Zhao
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan 250012, Shandong, China; and
| | - Lijuan Wang
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
21
|
Chen J, Liu M, Luo X, Peng L, Zhao Z, He C, He Y. Exosomal miRNA-486-5p derived from rheumatoid arthritis fibroblast-like synoviocytes induces osteoblast differentiation through the Tob1/BMP/Smad pathway. Biomater Sci 2020; 8:3430-3442. [PMID: 32406432 DOI: 10.1039/c9bm01761e] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathogenesis of rheumatoid arthritis (RA) is related to the inhibition of osteoblast differentiation. Exosomes secreted from RA fibroblast-like synoviocytes (RA-FLSs-exos) are associated with the pathogenesis of RA and microRNAs (miRNAs) being crucial for RA progression. Accordingly, the aim of the present study is to elucidate the effect of RA-FLS-derived exosomes on osteoblast differentiation and further identify exosomal cargos responsible for this effect. RA-FLSs were isolated from a RA patient and osteoblasts from the donor bone. Isolated RA-FLSs-exos were co-cultured with osteoblasts. Osteoblast differentiation was evaluated by ALP quantification assays, Alizarin Red S staining, and determining markers of osteoblast activity (Osx, OC, Col1a1 and Dlx2). Collagen induced arthritis (CIA)-induced mouse models were established. RA-FLSs-exo could be phagocytosed by osteoblasts. Elevating the expression of miR-486-5p in RA-FLSs-exo promoted osteoblast differentiation. miR-486-5p targeted Tob1 and activated the BMP/Smad signaling pathway in osteoblasts. In addition, RA-FLSs-exo containing miR-486-5p facilitated osteoblast differentiation by activating the BMP/Smad signaling pathway and repressing Tob1. Moreover, RA-FLSs-exo containing miR-486-5p alleviated the disease severity of RA by decreasing Tob1 expression in CIA-induced mice. To sum up, RA-FLSs-exo carrying miR-486-5p serve as a promoter for osteoblast differentiation in RA, ultimately highlighting a promising competitive new target for RA treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China.
| | | | | | | | | | | | | |
Collapse
|
22
|
Zhang G, Ferg M, Lübke L, Takamiya M, Beil T, Gourain V, Diotel N, Strähle U, Rastegar S. Bone morphogenetic protein signaling regulates Id1-mediated neural stem cell quiescence in the adult zebrafish brain via a phylogenetically conserved enhancer module. Stem Cells 2020; 38:875-889. [PMID: 32246536 DOI: 10.1002/stem.3182] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/08/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
In the telencephalon of adult zebrafish, the inhibitor of DNA binding 1 (id1) gene is expressed in radial glial cells (RGCs), behaving as neural stem cells (NSCs), during constitutive and regenerative neurogenesis. Id1 controls the balance between resting and proliferating states of RGCs by promoting quiescence. Here, we identified a phylogenetically conserved cis-regulatory module (CRM) mediating the specific expression of id1 in RGCs. Systematic deletion mapping and mutation of conserved transcription factor binding sites in stable transgenic zebrafish lines reveal that this CRM operates via conserved smad1/5 and 4 binding motifs under both homeostatic and regenerative conditions. Transcriptome analysis of injured and uninjured telencephala as well as pharmacological inhibition experiments identify a crucial role of bone morphogenetic protein (BMP) signaling for the function of the CRM. Our data highlight that BMP signals control id1 expression and thus NSC proliferation during constitutive and induced neurogenesis.
Collapse
Affiliation(s)
- Gaoqun Zhang
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Marco Ferg
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Luisa Lübke
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Masanari Takamiya
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Tanja Beil
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Victor Gourain
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Uwe Strähle
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
23
|
Chen XQ, Meng FQ, Xiong H, Wang YL, Tang WH, Zou YM. Identification of BTG1 Status in Solid Cancer for Future Researches Using a System Review and Meta-analysis. Curr Med Sci 2020; 40:85-94. [PMID: 32166669 DOI: 10.1007/s11596-020-2150-z] [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: 05/09/2019] [Revised: 12/10/2019] [Indexed: 12/24/2022]
Abstract
Abundant studies have been conducted to identify how B-cell translocation gene 1 protein (BTG1) gene affects the differentiation, proliferation, metastasis of cancer cells, and how it further regulates the generation or development of diseases to influence the prognosis of patients. However, the data from single research were not powerful enough. The correlations between BTG1 expression and mechanisms of tumorigenesis or prognosis of patients are still in controversial. Our system review and meta-analysis provided a complete explanation about the association between BTG1 expression and clinicopathological features or prognosis of patients, which further laid a foundation for future research on BTG1. Fifteen eligible studies consisting of 1992 participants were included. We uncovered that BTG1 expression in solid tumors was associated with lymph node status (RR=0.66, 95% CI: 0.58-0.75, P=0.142), TMN stage status (RR=2.13, 95% CI: 1.71-2.65, P=0.001), T category (RR=1.90, 95% CI: 1.20-3.00, P=0.000), histological differentiation (RR=1.91, 95% CI: 1.55-2.37, P=0.012), vascular invasion (RR=0.90, 95% CI: 0.57-1.41, P=0.001). BTG1 low expression was significantly associated with overall survival (OS) (HR=0.47, 95% CI: 0.38-0.67, P=0.000). It concluded that BTG1 possessed the potential value for future research and could be recommended as a significant biomarker in solid tumor.
Collapse
Affiliation(s)
- Xiu-Qiong Chen
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | | | - Hua Xiong
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ya-Li Wang
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Hua Tang
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan-Mei Zou
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
24
|
Jia B, Qiu X, Chen J, Sun X, Zheng X, Zhao J, Li Q, Wang Z. A feed-forward regulatory network lncPCAT1/miR-106a-5p/E2F5 regulates the osteogenic differentiation of periodontal ligament stem cells. J Cell Physiol 2019; 234:19523-19538. [PMID: 30997692 PMCID: PMC6767496 DOI: 10.1002/jcp.28550] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/15/2022]
Abstract
Periodontal ligament stem cells (PDLSCs) are characterized by multiple differentiation potential and potent self-renewal ability, yet much remains to be elucidated that what determines these properties. Long noncoding RNAs (lncRNAs) have been suggested to involve in multiple biological process under physiological and pathological conditions, including osteogenic differentiation. In the present study, we performed comprehensive lncRNA profiling by lncRNA microarray analysis and identified prostate cancer-associated ncRNA transcript-1 (lncPCAT1) was gradually increased in PDLSCs during consecutive osteogenic induction, and it could further positively regulate the osteogenic differentiation both in vitro and in vivo, whereas lncPCAT1 inhibition led to suppressed osteogenic differentiation. Thereafter, we inferred a predicted interaction between lncPCAT1 and miR-106a-5p and then confirmed the direct binding sites of miR-106a-5p on lncPCAT1. Although miR-106a-5p upregulation led to decreased osteogenic differentiation, lncPCAT1 overexpression could reverse its suppression, indicating that lncPCAT1 act as a competing endogenous RNA for miR-106a-5p. Moreover, lncPCAT1 could sponge miR-106a-5p to upregulate miR-106a-5p-targeted gene BMP2, which was a crucial gene involved in osteogenic differentiation. Interestingly, we found that E2F5, another target of miR-106a-5p, could bind to the promoter of lncPCAT1 and then form a feed-forward regulatory network targeting BMP2. In conclusion, our study provided a novel lncRNA-miRNA feed-forward regulatory network and a promising target to modulate the osteogenic differentiation of PDLSCs.
Collapse
Affiliation(s)
- Bo Jia
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaoling Qiu
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Jun Chen
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiang Sun
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Xianghuai Zheng
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Jianjiang Zhao
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Qin Li
- Department of Plastic SurgeryGuangzhou School of Clinical Medicine, Southern Medical University (Guangzhou General Hospital of Guangzhou Military Region)GuangzhouChina
| | - Zhiping Wang
- Department of Oral SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
25
|
LncRNA MSC-AS1 promotes osteogenic differentiation and alleviates osteoporosis through sponging microRNA-140–5p to upregulate BMP2. Biochem Biophys Res Commun 2019; 519:790-796. [DOI: 10.1016/j.bbrc.2019.09.058] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/14/2019] [Indexed: 12/27/2022]
|
26
|
Xiao J, Cao P, Wang C, Huang D, Lian M, Song Y, Yin W, Zheng K, Gu Z, Gu Y, Feng G, Feng X. The Forkhead Box C1, a Novel Negative Regulator of Osteogenesis, Plays a Crucial Role in Odontogenic Differentiation of Dental Pulp Stem Cells. Cell Reprogram 2019; 20:312-319. [PMID: 30277823 DOI: 10.1089/cell.2018.0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The forkhead box C1 (Foxc1) protein, a member of the forkhead/winged helix transcription factor family, is required in stem cell developmental processes. Recently, multiple studies have indicated the crucial role of Foxc1 in mesenchymal stem cell differentiation, but the precise effects and mechanisms on dental pulp stem cells (DPSCs) remain unclear. In this study, we evaluate the role of Foxc1 on the odontogenic differentiation and proliferation of DPSCs. Our results show that Foxc1 decreases time dependently in odontogenic differentiation of DPSCs. Meanwhile, overexpression of Foxc1 could significantly inhibit the mineralization of DPSCs and the expression of odontogenic-related genes, such as runt-related transcription factor 2 (Runx2), dentin sialophosphoprote (DSPP), and dentin matrix acidic phosphoprotein 1 (DMP-1). Foxc1 overexpression does not significantly alter the proliferation of DPSCs. In addition, Foxc1 reduces the expression of p-Smad1/5, an important modulator of bone morphogenetic protein (BMP)/Smad signaling pathway, inhibiting BMP/Smad signaling pathway. In conclusion, our data demonstrated that Foxc1 inhibits odontogenic differentiation of DPSCs and odontogenic-related gene expression through the BMP/Smad signaling pathway which may be useful for the dental regeneration and repair.
Collapse
Affiliation(s)
- Jingwen Xiao
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Peipei Cao
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Chenfei Wang
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Dan Huang
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Min Lian
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Yihua Song
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Weiwei Yin
- 2 Department of Stomatology, Stomatological Hospital of Nantong City , Nantong, Jiangsu, China
| | - Ke Zheng
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Zhifeng Gu
- 3 Department of Rheumatology, Affiliated Hospital of Nantong University , Nantong, Jiangsu, China
| | - Yongchun Gu
- 4 Department of Stomatology, First People's Hospital of Wujiang District, Nantong University , Suzhou, Jiangsu, China
| | - Guijuan Feng
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Xingmei Feng
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| |
Collapse
|
27
|
Moghaddam T, Neshati Z. Role of microRNAs in osteogenesis of stem cells. J Cell Biochem 2019; 120:14136-14155. [PMID: 31069839 DOI: 10.1002/jcb.28689] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/21/2022]
Abstract
Osteogenic differentiation is a controlled developmental process in which external and internal factors including cytokines, growth factors, transcription factors (TFs), signaling pathways and microRNAs (miRNAs) play important roles. Various stimulatory and inhibitory TFs contribute to osteogenic differentiation and are responsible for bone development. In addition, cross-talk between several complex signaling pathways regulates the osteogenic differentiation of some stem cells. Although much is known about regulatory genes and signaling pathways in osteogenesis, the role of miRNAs in osteogenic differentiation still needs to be explored. miRNAs are small, approximately 22 nucleotides, single-stranded nonprotein coding RNAs which are abundant in many mammalian cell types. They paly significant regulated roles in various biological processes and serve as promising biomarkers for disease states. Recently, emerging evidence have shown that miRNAs are the key regulators of osteogenesis of stem cells. They may endogenously regulate osteogenic differentiation of stem cells through direct targeting of positive or negative directors of osteogenesis and depending on the target result in the promotion or inhibition of osteogenic differentiation. This review aims to provide a general overview of miRNAs participating in osteogenic differentiation of stem cells and explain their regulatory effect based on the genes targeted with these miRNAs.
Collapse
Affiliation(s)
- Tayebe Moghaddam
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zeinab Neshati
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
28
|
Li J, Liu C, Li Y, Zheng Q, Xu Y, Liu B, Sun W, Li Y, Ji S, Liu M, Zhang J, Zhao D, Du R, Liu Z, Zhong G, Sun C, Wang Y, Song J, Zhang S, Qin J, Ling S, Wang X, Li Y. TMCO1-mediated Ca 2+ leak underlies osteoblast functions via CaMKII signaling. Nat Commun 2019; 10:1589. [PMID: 30962442 PMCID: PMC6453895 DOI: 10.1038/s41467-019-09653-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Transmembrane and coiled-coil domains 1 (TMCO1) is a recently identified Ca2+ leak channel in the endoplasmic reticulum. TMCO1 dysfunction in humans is associated with dysmorphism, mental retardation, glaucoma and the occurrence of cancer. Here we show an essential role of TMCO1 in osteogenesis mediated by local Ca2+/CaMKII signaling in osteoblasts. TMCO1 levels were significantly decreased in bone from both osteoporosis patients and bone-loss mouse models. Tmco1−/− mice exhibited loss of bone mass and altered microarchitecture characteristic of osteoporosis. In the absence of TMCO1, decreased HDAC4 phosphorylation resulted in nuclear enrichment of HADC4, which leads to deacetylation and degradation of RUNX2, the master regulator of osteogenesis. We further demonstrate that TMCO1-mediated Ca2+ leak provides local Ca2+ signals to activate the CaMKII-HDAC4-RUNX2 signaling axis. The establishment of TMCO1 as a pivotal player in osteogenesis uncovers a novel potential therapeutic target for ameliorating osteoporosis. TMCO1 is a recently described endoplasmic reticular Ca2+ channel. Here, the authors show it is important for osteoblast function and bone formation in mice, and identify a novel pathway linking local increases in Ca2+ at the ER surface with the posttranslational modification of RUNX2.
Collapse
Affiliation(s)
- Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, China
| | - Caizhi Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qiaoxia Zheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Youjia Xu
- The Second Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Beibei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Weijia Sun
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yuan Li
- The Second Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Shuhui Ji
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jing Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Ruikai Du
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Cuiwei Sun
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Yanqing Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Jinping Song
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China.
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| |
Collapse
|
29
|
Rakmanee T, Calciolari E, Olsen I, Darbar U, Griffiths GS, Petrie A, Donos N. Expression of growth mediators in the gingival crevicular fluid of patients with aggressive periodontitis undergoing periodontal surgery. Clin Oral Investig 2018; 23:3307-3318. [DOI: 10.1007/s00784-018-2752-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
|
30
|
Liu X, Cao F, Liu S, Mi Y, Liu J. BMP2/Smad signaling pathway is involved in the inhibition function of fibroblast growth factor 21 on vascular calcification. Biochem Biophys Res Commun 2018; 503:930-937. [PMID: 29932916 DOI: 10.1016/j.bbrc.2018.06.098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 06/18/2018] [Indexed: 11/17/2022]
Abstract
Vascular calcification is extremely common and associated with major adverse cardiovascular events. Fibroblast growth factor (FGF) 21 has been identified as a potent metabolic regulator and a protector of the cardiovascular system. In this study, we aimed to investigate the effect of FGF21 on calcification of vascular smooth muscle cell (VSMC) and its mechanism. FGF21 inhibited beta-glycerophosphate (BGP) induced mineralization in VSMCs as determined by calcium concentration and Alizarin Red S. FGF21 suppressed BGP-induced BMP2/Smad signaling pathway components as well as osteoblast differentiation markers. FGF21 and Noggin could synergistically inhibit BGP-induced BMP2/Smad pathway expressions and calcification. Taken together, FGF21 inhibits vascular calcification in vitro by modulating BMP2/Smad signaling pathway.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Fangying Cao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuang Liu
- Department of Pulmonary and Critical Care Medicine, Peking University International Hospital, Beijing, China
| | - Yuhong Mi
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jinghua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
31
|
Wang D, Jiang X, Lu A, Tu M, Huang W, Huang P. BMP14 induces tenogenic differentiation of bone marrow mesenchymal stem cells in vitro. Exp Ther Med 2018; 16:1165-1174. [PMID: 30116367 PMCID: PMC6090266 DOI: 10.3892/etm.2018.6293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/24/2018] [Indexed: 01/28/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are pluripotent cells, which have the capacity to differentiate into various types of mesenchymal cell phenotypes, including osteoblasts, chondroblasts, myoblasts and tendon fibroblasts (TFs). The molecular mechanism for tenogenic differentiation of BMSCs is still unknown. The present study investigated the effects of bone morphogenetic protein (BMP) 14 on BMSC differentiation in vitro. It was revealed that BMP14 significantly increased the expression of tendon markers (scleraxis and tenomodulin) at the mRNA and protein level, which led to the upregulation of sirtuin 1 (Sirt1) expression. The gain or loss of Sirt1 function may promote or inhibit tenogenic differentiation by deacetylating the peroxisome proliferator-activated receptor (PPAR)-γ. BMP14 also triggered the phosphorylation of c-Jun N-terminal kinase (JNK) and Smad1; overexpression of Sirt1 significantly increased the phosphorylation and knockdown of Sirt1 significantly decreased the phosphorylation. The inhibition of JNK and Smad significantly increased the acetylation of PPARγ and inhibited the expression of tenogenic differentiation markers. These results suggest that BMP14 may induce the tenogenic differentiation of BMSCs via the Sirt1-JNK/Smad1-PPARγ signaling pathway. The present study provided a cellular and molecular basis for the development of novel therapeutic strategies for tendon healing.
Collapse
Affiliation(s)
- Dan Wang
- Department of Orthopedics, Jinmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China.,Department of Orthopedics, Jingchu Center Hospital Affiliated to The Institute of Technology, Jingmen, Hubei 448000, P.R. China
| | - Xinhao Jiang
- Department of Orthopedics, Jinmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China.,Department of Orthopedics, Jingchu Center Hospital Affiliated to The Institute of Technology, Jingmen, Hubei 448000, P.R. China
| | - Aiqing Lu
- Department of Orthopedics, Jinmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China.,Department of Orthopedics, Jingchu Center Hospital Affiliated to The Institute of Technology, Jingmen, Hubei 448000, P.R. China
| | - Min Tu
- Department of Orthopedics, Jinmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China.,Department of Orthopedics, Jingchu Center Hospital Affiliated to The Institute of Technology, Jingmen, Hubei 448000, P.R. China
| | - Wei Huang
- Department of Orthopedics, Jinmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China.,Department of Orthopedics, Jingchu Center Hospital Affiliated to The Institute of Technology, Jingmen, Hubei 448000, P.R. China
| | - Ping Huang
- Department of Orthopedics, Jinmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China.,Department of Orthopedics, Jingchu Center Hospital Affiliated to The Institute of Technology, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
32
|
Klar RM. The Induction of Bone Formation: The Translation Enigma. Front Bioeng Biotechnol 2018; 6:74. [PMID: 29938204 PMCID: PMC6002665 DOI: 10.3389/fbioe.2018.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/22/2018] [Indexed: 11/25/2022] Open
Abstract
A paradigmatic shift in the way of thinking is what bone tissue engineering science requires to decrypt the translation conundrum from animal models into human. The deductive work of Urist (1965), who discerned the principle of bone induction from the pioneering works of Senn, Huggins, Lacroix, Levander, and other bone regenerative scientists, provided the basis that has assisted future bone tissue regenerative scientists to extend the bone tissue engineering field and its potential uses for bone regenerative medicine in humans. However, major challenges remain that are preventing the formation of bone by induction clinically. Growing experimental evidence is indicating that bone inductive studies are non-translatable from animal models into a clinical environment. This is preventing bone tissue engineering from reaching the next phase in development. Countless studies are trying to discern how the formation of bone by induction functions mechanistically, so as to try and solve this enigmatic problem. However, are the correct questions being asked? Why do bone inductive animal studies not translate into humans? Why do bone induction principles not yield the same extent of bone formation as an autogenous bone graft? What are bone tissue engineering scientists missing? By critically re-assessing the past and present discoveries of the bone induction field, this review article attempts to re-discover the field of bone formation by induction, identifying some key features that may have been missed. These include a detailed library of all proteins in bones and their arrangement in the 3D superstructure of the bone together with some other important criteria not considered by tissue engineering scientists. The review therefore not only re-iterates possible avenues of research that need to be re-explored but also seeks to guide present and future scientists in how they assess their own research in light of experimental design and results. By addressing these issues bone formation by induction without autografts might finally become clinically viable.
Collapse
Affiliation(s)
- Roland M. Klar
- Laboratory of Biomechanics and Experimental Orthopaedics, Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany
| |
Collapse
|
33
|
Sun J, Ermann J, Niu N, Yan G, Yang Y, Shi Y, Zou W. Histone demethylase LSD1 regulates bone mass by controlling WNT7B and BMP2 signaling in osteoblasts. Bone Res 2018; 6:14. [PMID: 29707403 PMCID: PMC5916912 DOI: 10.1038/s41413-018-0015-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/03/2018] [Accepted: 03/21/2018] [Indexed: 12/22/2022] Open
Abstract
Multiple regulatory mechanisms control osteoblast differentiation and function to ensure unperturbed skeletal formation and remodeling. In this study we identify histone lysine-specific demethylase 1(LSD1/KDM1A) as a key epigenetic regulator of osteoblast differentiation. Knockdown of LSD1 promoted osteoblast differentiation of human mesenchymal stem cells (hMSCs) in vitro and mice lacking LSD1 in mesenchymal cells displayed increased bone mass secondary to accelerated osteoblast differentiation. Mechanistic in vitro studies revealed that LSD1 epigenetically regulates the expression of WNT7B and BMP2. LSD1 deficiency resulted in increased BMP2 and WNT7B expression in osteoblasts and enhanced bone formation, while downregulation of WNT7B- and BMP2-related signaling using genetic mouse model or small-molecule inhibitors attenuated bone phenotype in vivo. Furthermore, the LSD1 inhibitor tranylcypromine (TCP) could increase bone mass in mice. These data identify LSD1 as a novel regulator of osteoblast activity and suggest LSD1 inhibition as a potential therapeutic target for treatment of osteoporosis.
Collapse
Affiliation(s)
- Jun Sun
- 1State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Joerg Ermann
- 2Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Ningning Niu
- 1State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Guang Yan
- 1State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Yang Yang
- 1State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| | - Yujiang Shi
- 3Newborn Medicine Division, Boston Children's Hospital and Department of Cell Biology, Harvard Medical School, Boston, MA 02115 USA
| | - Weiguo Zou
- 1State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031 China
| |
Collapse
|
34
|
Luo Y, Cao X, Chen J, Gu J, Zhao J, Sun J. MicroRNA‐224 suppresses osteoblast differentiation by inhibiting
SMAD4. J Cell Physiol 2018; 233:6929-6937. [PMID: 29693254 DOI: 10.1002/jcp.26596] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/12/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Yuan Luo
- Department of Orthopedic The First Affiliated Hospital of Soochow University Souzhou Jiangsu China
- Department of Orthopedic Taicang Affiliated Hospital of Soochow University Taicang Jiangsu China
| | - Xiaodong Cao
- Department of Orthopedic Taicang Affiliated Hospital of Soochow University Taicang Jiangsu China
| | - Junfeng Chen
- Department of Orthopedic Taicang Affiliated Hospital of Soochow University Taicang Jiangsu China
| | - Jianwei Gu
- Department of Orthopedic Taicang Affiliated Hospital of Soochow University Taicang Jiangsu China
| | - Jitong Zhao
- Department of Orthopedic Taicang Affiliated Hospital of Soochow University Taicang Jiangsu China
| | - Junying Sun
- Department of Orthopedic The First Affiliated Hospital of Soochow University Souzhou Jiangsu China
| |
Collapse
|
35
|
Ampuja M, Kallioniemi A. Transcription factors-Intricate players of the bone morphogenetic protein signaling pathway. Genes Chromosomes Cancer 2017; 57:3-11. [DOI: 10.1002/gcc.22502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- M. Ampuja
- BioMediTech Institute and Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
| | - Anne Kallioniemi
- BioMediTech Institute and Faculty of Medicine and Life Sciences; University of Tampere; Tampere Finland
- Fimlab Laboratories; Tampere Finland
| |
Collapse
|
36
|
Abstract
Inhibitory Smads (I-Smads) have conserved carboxy-terminal MH2 domains but highly divergent amino-terminal regions when compared with receptor-regulated Smads (R-Smads) and common-partner Smads (co-Smads). Smad6 preferentially inhibits Smad signaling initiated by the bone morphogenetic protein (BMP) type I receptors ALK-3 and ALK-6, whereas Smad7 inhibits both transforming growth factor β (TGF-β)- and BMP-induced Smad signaling. I-Smads also regulate some non-Smad signaling pathways. Here, we discuss the vertebrate I-Smads, their roles as inhibitors of Smad activation and regulators of receptor stability, as scaffolds for non-Smad signaling, and their possible roles in the nucleus. We also discuss the posttranslational modification of I-Smads, including phosphorylation, ubiquitylation, acetylation, and methylation.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
37
|
Maeda Y, Farina NH, Matzelle MM, Fanning PJ, Lian JB, Gravallese EM. Synovium-Derived MicroRNAs Regulate Bone Pathways in Rheumatoid Arthritis. J Bone Miner Res 2017; 32:461-472. [PMID: 27676131 PMCID: PMC5340607 DOI: 10.1002/jbmr.3005] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/19/2016] [Accepted: 09/24/2016] [Indexed: 12/11/2022]
Abstract
Articular bone erosion in rheumatoid arthritis (RA) is mediated by the interaction between inflammation and pathways regulating bone metabolism. Inflammation promotes osteoclastogenesis and also inhibits osteoblast function, further contributing to the persistence of erosions. MicroRNAs (miRNAs) are important regulators of skeletal remodeling and play a role in RA pathogenesis. We therefore determined the expression of miRNAs in inflamed synovial tissue and the role they play in pathways regulating osteoblast and osteoclast function. Using the serum transfer mouse model of RA in C57BL/6 mice, we performed Fluidigm high-throughput qPCR-based screening of miRNAs from nonarthritic and arthritic mice. Global gene expression profiling was also performed on Affymetrix microarrays from these same synovial samples. miRNA and mRNA expression profiles were subjected to comparative bioinformatics. A total of 536 upregulated genes and 417 downregulated genes were identified that are predicted targets of miRNAs with reciprocal expression changes. Gene ontology analysis of these genes revealed significant enrichment in skeletal pathways. Of the 22 miRNAs whose expression was most significantly changed (p < 0.01) between nonarthritic and arthritic mice, we identified their targets that both inhibit and promote bone formation. These miRNAs are predicted to target Wnt and BMP signaling pathway components. We validated miRNA array findings and demonstrated that secretion of miR-221-3p in exosomes was upregulated by synovial fibroblasts treated with the proinflammatory cytokine TNF. Overexpression of miR-221-3p suppressed calvarial osteoblast differentiation and mineralization in vitro. These results suggest that miRNAs derived from inflamed synovial tissues may regulate signaling pathways at erosion sites that affect bone loss and potentially also compensatory bone formation. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yukiko Maeda
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Nicholas H Farina
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | - Melissa M Matzelle
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Paul J Fanning
- Department of Orthopedic Surgery and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jane B Lian
- Department of Biochemistry, University of Vermont, Burlington, VT, USA.,Department of Orthopedic Surgery and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ellen M Gravallese
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
38
|
SMURF2 regulates bone homeostasis by disrupting SMAD3 interaction with vitamin D receptor in osteoblasts. Nat Commun 2017; 8:14570. [PMID: 28216630 PMCID: PMC5321737 DOI: 10.1038/ncomms14570] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 01/10/2017] [Indexed: 01/17/2023] Open
Abstract
Coordination between osteoblasts and osteoclasts is required for bone health and homeostasis. Here we show that mice deficient in SMURF2 have severe osteoporosis in vivo. This low bone mass phenotype is accompanied by a pronounced increase in osteoclast numbers, although Smurf2-deficient osteoclasts have no intrinsic alterations in activity. Smurf2-deficient osteoblasts display increased expression of RANKL, the central osteoclastogenic cytokine. Mechanistically, SMURF2 regulates RANKL expression by disrupting the interaction between SMAD3 and vitamin D receptor by altering SMAD3 ubiquitination. Selective deletion of Smurf2 in the osteoblast lineage recapitulates the phenotype of germline Smurf2-deficient mice, indicating that SMURF2 regulates osteoblast-dependent osteoclast activity rather than directly affecting the osteoclast. Our results reveal SMURF2 as an important regulator of the critical communication between osteoblasts and osteoclasts. Furthermore, the bone mass phenotype in Smurf2- and Smurf1-deficient mice is opposite, indicating that SMURF2 has a non-overlapping and, in some respects, opposite function to SMURF1. The balance between osteoclast and osteoblast-mediated bone turnover is essential for bone health and homeostasis. Here the authors show that both germline and osteoblast-specific Smurf2-deficient mice have osteoporosis as a result of increased osteoblast RANKL production and excess osteoclastogenesis.
Collapse
|
39
|
Wu Q, Fang T, Lang H, Chen M, Shi P, Pang X, Qi G. Comparison of the proliferation, migration and angiogenic properties of human amniotic epithelial and mesenchymal stem cells and their effects on endothelial cells. Int J Mol Med 2017; 39:918-926. [PMID: 28259958 PMCID: PMC5360425 DOI: 10.3892/ijmm.2017.2897] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
In vivo studies have shown that amnion-produced growth factors participate in many diseases that involve angiogenesis, re-epithelialization and immunomodulation. Although human amniotic epithelial cells (hAECs) and human amniotic mesenchymal stem cells (hAMSCs) can be obtained from amniotic membranes, there is little information regarding their biological differences. The aim of the present study was to isolate and characterize cells from human amnions, to investigate the biological potential and behavior of these cells on the function of endothelial cells in vivo and in vitro and to examine variations in the expression profile of growth factors in different human amnion-derived cell types. Amnion fragments were enzymatically digested into two cell fractions, which were analyzed by mesenchymal and epithelial cell markers. Human aortic endothelial cells (hAoECs) were cultured with conditioned medium (CdM) collected from hAECs or hAMSCs. We used scratch and Transwell assays to evaluate migration ability; Cell Counting Kit-8 (CCK-8) and cell cycle analysis to evaluate proliferation ability; and a Matrigel tube formation assay to evaluate angiogenesis ability. To detect expression of angiogenesis-related genes, qPCR and enzyme-linked immunosorbent assay (ELISA) analyses were conducted. As stem cells, hAECs and hAMSCs all expressed the stem cell markers SSEA-4, OCT-4 and SOX-2. CdM obtained from hAECs promoted cell migration; CdM obtained from hAMSCs promoted cell proliferation; CdM obtained from hAECs and hAMSCs both promoted angiogenesis in hAoECs. Amnion-derived cells secreted significant amounts of angiogenic factors including HGF, IGF-1, VEGF, EGF, HB-EGF and bFGF, although differences in the cellular expression profile of these soluble factors were observed. Our results highlight that human amniotic epithelial and mesenchymal stem cells, which showed differences in their soluble factor secretion and angiogenic functions, could be ideal cell sources for regenerative medicine.
Collapse
Affiliation(s)
- Qianqian Wu
- Department of Geriatric Cardiology and Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Tao Fang
- Department of Orthopedic Surgery, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongxin Lang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Chinese Ministry of Public Health and Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang, Liaoning 110013, P.R. China
| | - Min Chen
- Department of Geriatric Cardiology and Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ping Shi
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xining Pang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Chinese Ministry of Public Health and Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang, Liaoning 110013, P.R. China
| | - Guoxian Qi
- Department of Geriatric Cardiology and Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
40
|
RNA-binding protein SAMD4 regulates skeleton development through translational inhibition of Mig6 expression. Cell Discov 2017; 3:16050. [PMID: 28163927 PMCID: PMC5259697 DOI: 10.1038/celldisc.2016.50] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022] Open
Abstract
Protein translation regulation has essential roles in inflammatory responses, cancer initiation and the pathogenesis of several neurodegenerative disorders. However, the role of the regulation of protein translation in mammalian skeleton development has been rarely elaborated. Here we report that the lack of the RNA-binding protein sterile alpha motif domain containing protein 4 (SAMD4) resulted in multiple developmental defects in mice, including delayed bone development and decreased osteogenesis. Samd4-deficient mesenchymal progenitors exhibit impaired osteoblast differentiation and function. Mechanism study demonstrates that SAMD4 binds the Mig6 mRNA and inhibits MIG6 protein synthesis. Consistent with this, Samd4-deficient cells have increased MIG6 protein level and knockdown of Mig6 rescues the impaired osteogenesis in Samd4-deficient cells. Furthermore, Samd4-deficient mice also display chondrocyte defects, which is consistent with the regulation of MIG6 protein level by SAMD4. These findings define SAMD4 as a previously unreported key regulator of osteoblastogenesis and bone development, implying that regulation of protein translation is an important mechanism governing skeletogenesis and that control of protein translation could have therapeutic potential in metabolic bone diseases, such as osteoporosis.
Collapse
|
41
|
Yang X, Huo H, Xiu C, Song M, Han Y, Li Y, Zhu Y. Inhibition of osteoblast differentiation by aluminum trichloride exposure is associated with inhibition of BMP-2/Smad pathway component expression. Food Chem Toxicol 2016; 97:120-126. [PMID: 27600293 DOI: 10.1016/j.fct.2016.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 02/07/2023]
Abstract
Bone morphogenetic protein-2 (BMP-2)/Smad signaling pathway plays an important role in regulating osteoblast (OB) differentiation. OB differentiation is a key process of bone formation. Aluminum (Al) exposure inhibits bone formation and causes Al-induced bone disease. However, the mechanism is not fully understood. To investigate whether BMP-2/Smad signaling pathway is associated with OB differentiation in aluminum trichloride (AlCl3)-treated OBs, the primary rat OBs were cultured and exposed to 0 (control group, CG), 1/40 IC50 (low-dose group, LG), 1/20 IC50 (mid-dose group, MG), and 1/10 IC50 (high-dose group, HG) of AlCl3 for 24 h, respectively. We found that the expressions of OB differentiation markers (Runx-2, Osterix and ALP) and BMP-2/Smad signaling pathway components (BMP-2, BMPR-IA, p-BMPR-IA, BMPR-II, p-Smad1/5/8 and p-Smad1/5/8/4) were all decreased in AlCl3-treated OBs compared with the CG. These results indicated that inhibition of OB differentiation by AlCl3 was associated with inhibition of BMP-2/Smad pathway component expression. Our findings provide a novel insight into the mechanism of AlCl3-induced bone disease.
Collapse
Affiliation(s)
- Xu Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Hui Huo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Chunyu Xiu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Miao Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yanfei Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yanfei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| | - Yanzhu Zhu
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun 130112, China.
| |
Collapse
|
42
|
Didonna A, Cekanaviciute E, Oksenberg JR, Baranzini SE. Immune cell-specific transcriptional profiling highlights distinct molecular pathways controlled by Tob1 upon experimental autoimmune encephalomyelitis. Sci Rep 2016; 6:31603. [PMID: 27546286 PMCID: PMC4992865 DOI: 10.1038/srep31603] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/20/2016] [Indexed: 11/09/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system characterized by focal lymphocytic infiltration, demyelination and neurodegeneration. Despite the recent advances in understanding MS molecular basis, no reliable biomarkers have been identified yet to monitor disease progression. Our group has previously reported that low levels of TOB1 in CD4(+) T cells are strongly associated with a higher risk of MS conversion in individuals experiencing an initial demyelinating event. Consistently, Tob1 ablation in mice exacerbates the clinical phenotype of the MS model experimental autoimmune encephalomyelitis (EAE). To shed light on Tob1 molecular functions in the immune system, we have conducted the first cell-based transcriptomic analysis in Tob1(-/-) and wildtype mice upon EAE. Next-generation sequencing was employed to characterize the changes in gene expression in T and B cells at pre- and post-symptomatic EAE stages. Remarkably, we found only modest overlap among the different genetic signatures, suggesting that Tob1 may control distinct genetic programs in the different cytotypes. This hypothesis was corroborated by gene ontology and global interactome analyses, which highlighted specific cellular pathways in each cellular subset before and after EAE induction. In summary, our work pinpoints a multifaceted activity of Tob1 in both homeostasis and disease progression.
Collapse
Affiliation(s)
- Alessandro Didonna
- Department of Neurology, University of California San Francisco, San Francisco, California 94158, USA
| | - Egle Cekanaviciute
- Department of Neurology, University of California San Francisco, San Francisco, California 94158, USA
| | - Jorge R Oksenberg
- Department of Neurology, University of California San Francisco, San Francisco, California 94158, USA
| | - Sergio E Baranzini
- Department of Neurology, University of California San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
43
|
Fang T, Wu Q, Mu S, Yang L, Liu S, Fu Q. Shikonin stimulates MC3T3-E1 cell proliferation and differentiation via the BMP-2/Smad5 signal transduction pathway. Mol Med Rep 2016; 14:1269-74. [DOI: 10.3892/mmr.2016.5363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 05/10/2016] [Indexed: 11/06/2022] Open
|
44
|
Ali IHA, Brazil DP. Bone morphogenetic proteins and their antagonists: current and emerging clinical uses. Br J Pharmacol 2016; 171:3620-32. [PMID: 24758361 DOI: 10.1111/bph.12724] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/02/2014] [Accepted: 04/08/2014] [Indexed: 12/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the TGFβ superfamily of secreted cysteine knot proteins that includes TGFβ1, nodal, activins and inhibins. BMPs were first discovered by Urist in the 1960s when he showed that implantation of demineralized bone into intramuscular tissue of rabbits induced bone and cartilage formation. Since this seminal discovery, BMPs have also been shown to play key roles in several other biological processes, including limb, kidney, skin, hair and neuronal development, as well as maintaining vascular homeostasis. The multifunctional effects of BMPs make them attractive targets for the treatment of several pathologies, including bone disorders, kidney and lung fibrosis, and cancer. This review will summarize current knowledge on the BMP signalling pathway and critically evaluate the potential of recombinant BMPs as pharmacological agents for the treatment of bone repair and tissue fibrosis in patients.
Collapse
Affiliation(s)
- Imran H A Ali
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
| | | |
Collapse
|
45
|
Wu M, Chen G, Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res 2016; 4:16009. [PMID: 27563484 PMCID: PMC4985055 DOI: 10.1038/boneres.2016.9] [Citation(s) in RCA: 1035] [Impact Index Per Article: 129.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) and bone morphogenic protein (BMP) signaling has fundamental roles in both embryonic skeletal development and postnatal bone homeostasis. TGF-βs and BMPs, acting on a tetrameric receptor complex, transduce signals to both the canonical Smad-dependent signaling pathway (that is, TGF-β/BMP ligands, receptors, and Smads) and the non-canonical-Smad-independent signaling pathway (that is, p38 mitogen-activated protein kinase/p38 MAPK) to regulate mesenchymal stem cell differentiation during skeletal development, bone formation and bone homeostasis. Both the Smad and p38 MAPK signaling pathways converge at transcription factors, for example, Runx2 to promote osteoblast differentiation and chondrocyte differentiation from mesenchymal precursor cells. TGF-β and BMP signaling is controlled by multiple factors, including the ubiquitin–proteasome system, epigenetic factors, and microRNA. Dysregulated TGF-β and BMP signaling result in a number of bone disorders in humans. Knockout or mutation of TGF-β and BMP signaling-related genes in mice leads to bone abnormalities of varying severity, which enable a better understanding of TGF-β/BMP signaling in bone and the signaling networks underlying osteoblast differentiation and bone formation. There is also crosstalk between TGF-β/BMP signaling and several critical cytokines’ signaling pathways (for example, Wnt, Hedgehog, Notch, PTHrP, and FGF) to coordinate osteogenesis, skeletal development, and bone homeostasis. This review summarizes the recent advances in our understanding of TGF-β/BMP signaling in osteoblast differentiation, chondrocyte differentiation, skeletal development, cartilage formation, bone formation, bone homeostasis, and related human bone diseases caused by the disruption of TGF-β/BMP signaling.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| | - Guiqian Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA; Department of neurology, Bruke Medical Research Institute, Weil Cornell Medicine of Cornell University, White Plains, USA
| | - Yi-Ping Li
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| |
Collapse
|
46
|
Bone morphogenetic protein signaling in musculoskeletal cancer. J Cancer Res Clin Oncol 2016; 142:2061-72. [PMID: 27043154 DOI: 10.1007/s00432-016-2149-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 03/17/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE Bone morphogenetic proteins (BMPs) belong to the transforming growth factor-β (TGF-β) superfamily of proteins; they were initially named after their ability to induce ectopic bone formation. Published studies have proved BMPs' role in a variety of biological processes such as embryogenesis and patterning of body axes, and maintaining adult tissue homeostasis. Other studies have focused on BMPs properties, functions and possible involvement in skeletal diseases, including cancer. METHODS A literature search mainly paying attention to the role of BMPs in musculoskeletal tumors was performed in electronic databases. RESULTS This article discusses BMPs synthesis and signaling, and summarizes their prominent roles in the skeletal system for the differentiation of osteoblasts, osteocytes and chondrocytes. CONCLUSIONS The review emphasizes on the role of BMP signaling in the initiation and progression of musculoskeletal cancer.
Collapse
|
47
|
Si K, Kandel ER. The Role of Functional Prion-Like Proteins in the Persistence of Memory. Cold Spring Harb Perspect Biol 2016; 8:a021774. [PMID: 27037416 DOI: 10.1101/cshperspect.a021774] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Prions are a self-templating amyloidogenic state of normal cellular proteins, such as prion protein (PrP). They have been identified as the pathogenic agents, contributing to a number of diseases of the nervous system. However, the discovery that the neuronal RNA-binding protein, cytoplasmic polyadenylation element-binding protein (CPEB), has a prion-like state that is involved in the stabilization of memory raised the possibility that prion-like proteins can serve normal physiological functions in the nervous system. Here, we review recent experimental evidence of prion-like properties of neuronal CPEB in various organisms and propose a model of how the prion-like state may stabilize memory.
Collapse
Affiliation(s)
- Kausik Si
- Stowers Institute for Medical Research, Kansas City, Missouri 64113 Department of Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Eric R Kandel
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815-6789 Departments of Neuroscience and Psychiatry, College of Physicians and Surgeons of Columbia University, New York, New York 10027 Zuckerman Mind Brain Behavior Institute, New York State Psychiatric Institute, New York, New York 10032 Kavli Institute for Brain Sciences, New York, New York 10032
| |
Collapse
|
48
|
Wang C, Yu T, Tan L, Cheng J. Bioinformatics analysis of gene expression profile in callus tissues of osteoporotic phenotype mice induced by osteoblast-specific Krm2 overexpression. Int J Rheum Dis 2016; 19:1263-1271. [PMID: 26929007 DOI: 10.1111/1756-185x.12840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE The aim of this study was to explore the molecular mechanism of fracture healing in osteoporotic mice. METHODS The gene expression profiles of callus tissues of osteoporotic mice and controls were obtained from Gene Expression Omnibus database. The differentially expressed genes (DEGs) and their related biological function and pathways were investigated. In addition, the protein-protein interaction (PPI) network was constructed for DEG encoding proteins and the differentially expressed transcriptional factor was screened. RESULTS There were 275 up-regulated genes and 347 down-regulated genes. The collagen metabolic process biological function was significantly enriched by down-regulated genes. Extracellular matrix (ECM)-receptor interaction was a significant pathway that was enriched by differentially expressed genes. In PPI (protein-protein interaction) network, Pcna was the significant node with highest connective degrees. Other hub nodes, such as Ccnb2 and Rrm2, were closely associated with the p53 signaling pathway. Tal1 and Smad6 were found to be differentially expressed transcription factors. CONCLUSION The dysregulated collagen metabolic process, ECM-receptor interaction and p53 signaling pathway may be responsible for impaired fracture healing of osteoporotic mice. The hub nodes (such as Ccnb2 and Rrm2) and differentially expressed TFs (such as Tal1 and Smad6) play a critical role in bone remodeling of osteoporotic individuals.
Collapse
Affiliation(s)
- Chengxue Wang
- Department of Trauma, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Tiecheng Yu
- Department of Trauma, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lei Tan
- Department of Trauma, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jieping Cheng
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
49
|
Chan SCW, Tekari A, Benneker LM, Heini PF, Gantenbein B. Osteogenic differentiation of bone marrow stromal cells is hindered by the presence of intervertebral disc cells. Arthritis Res Ther 2015; 18:29. [PMID: 26809343 PMCID: PMC4727301 DOI: 10.1186/s13075-015-0900-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/14/2015] [Indexed: 02/04/2023] Open
Abstract
Background Clinical observations indicate that the presence of nucleus pulposus (NP) tissue during spinal fusion hinders the rate of disc ossification. While the underlying mechanism remains unknown, this observation could be due to incomplete removal of NP cells (NPCs) that secrete factors preventing disc calcification, such as bone morphogenetic protein (BMP) antagonists including noggin and members of the DAN (differential screening selected gene aberrative in neuroblastoma) family. Methods Monolayer human bone marrow-derived mesenchymal stem cells (MSCs) were cocultured withNPCs and annulus fibrosus cells (AFCs) embedded in alginate for 21 days. At the end of coculture, MSCs were stained for mineral deposition by alizarin red, and relative expression of bone-related genes [Runt-related transcription factor 2, (RUNX2), Osteopontin (OPN), and Alkaline phosphatase (ALP)] and ALP activity were analyzed. Relative expression of three BMP antagonists, chordin (CHRD), gremlin (GREM1), and noggin (NOG), was determined in primary human NPCs and AFCs. These cells were also stained for Gremlin and Noggin by immunocytochemistry. Results Alizarin red staining showed that MSC osteogenesis in monolayer cultures was inhibited by coculture with NPCs or AFCs. ALP activity and RT-PCR analyses confirmed these results and demonstrated inhibition of osteogenesis of MSC in the presence of disc cells. NOG was significantly up-regulated in MSCs after coculture. Relative gene expression of intervertebral disc (IVD) cells showed higher expression of GREM1 in NPCs than in AFCs. Conclusions We show that primary IVD cells inhibit osteogenesis of MSCs. BMP inhibitors NOG, GREM1 and CHRD were expressed in IVD cells. GREM1 appears to be differentially expressed in NPCs and AFCs. Our results have implications for the design and development of treatments for non-union in spinal fusion.
Collapse
Affiliation(s)
- Samantha C W Chan
- Tissue and Organ Mechanobiology, Institute for Surgical Technology and Biomechanics, University of Bern, Stauffacherstrasse 78, Bern, CH-3014, Switzerland. .,Biointerfaces, EMPA, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, St Gallen, CH-9014, Switzerland.
| | - Adel Tekari
- Tissue and Organ Mechanobiology, Institute for Surgical Technology and Biomechanics, University of Bern, Stauffacherstrasse 78, Bern, CH-3014, Switzerland.
| | - Lorin M Benneker
- Department for Orthopedic Surgery and Traumatology, Inselspital, University of Bern, Freiburgstrasse 4, Bern, CH-3010, Switzerland. .,AOSpine Research Network, Stettbachstrasse 6, Dübendorf, CH-8600, Switzerland.
| | - Paul F Heini
- Orthopedic Department, Sonnenhof Clinic, Buchserstrasse 30, Bern, CH-3006, Switzerland.
| | - Benjamin Gantenbein
- Tissue and Organ Mechanobiology, Institute for Surgical Technology and Biomechanics, University of Bern, Stauffacherstrasse 78, Bern, CH-3014, Switzerland. .,AOSpine Research Network, Stettbachstrasse 6, Dübendorf, CH-8600, Switzerland.
| |
Collapse
|
50
|
Takahashi A, Adachi S, Morita M, Tokumasu M, Natsume T, Suzuki T, Yamamoto T. Post-transcriptional Stabilization of Ucp1 mRNA Protects Mice from Diet-Induced Obesity. Cell Rep 2015; 13:2756-67. [PMID: 26711342 DOI: 10.1016/j.celrep.2015.11.056] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 10/10/2015] [Accepted: 11/17/2015] [Indexed: 11/24/2022] Open
Abstract
Uncoupling protein 1 (Ucp1) contributes to thermogenesis, and its expression is regulated at the transcriptional level. Here, we show that Ucp1 expression is also regulated post-transcriptionally. In inguinal white adipose tissue (iWAT) of mice fed a high-fat diet (HFD), Ucp1 level decreases concomitantly with increases in Cnot7 and its interacting partner Tob. HFD-fed mice lacking Cnot7 and Tob express elevated levels of Ucp1 mRNA in iWAT and are resistant to diet-induced obesity. Ucp1 mRNA has an elongated poly(A) tail and persists in iWAT of Cnot7(-/-) and/or Tob(-/-) mice on a HFD. Ucp1 3'-UTR-containing mRNA is more stable in cells expressing mutant Tob that is unable to bind Cnot7 than in WT Tob-expressing cells. Tob interacts with BRF1, which binds to an AU-rich element in the Ucp1 3'-UTR. BRF1 knockdown partially restores the stability of Ucp1 3'-UTR-containing mRNA. Thus, the Cnot7-Tob-BRF1 axis inhibits Ucp1 expression and contributes to obesity.
Collapse
Affiliation(s)
- Akinori Takahashi
- Cell Signal Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa 904-0412, Japan
| | - Shungo Adachi
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Masahiro Morita
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada
| | - Miho Tokumasu
- Cell Signal Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa 904-0412, Japan
| | - Tohru Natsume
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Toru Suzuki
- Cell Signal Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa 904-0412, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology, Kunigami, Okinawa 904-0412, Japan.
| |
Collapse
|