1
|
Zhao H, Tu X. The potential key genes within focal adhesion that regulate mesenchymal stem cells osteogenesis or adipogenesis in microgravity related disuse osteoporosis: an integrated analysis. Front Endocrinol (Lausanne) 2025; 16:1469400. [PMID: 40130165 PMCID: PMC11930814 DOI: 10.3389/fendo.2025.1469400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/14/2025] [Indexed: 03/26/2025] Open
Abstract
This study aimed to identify key genes related to focal adhesions (FA) and cells involved in osteoblast (OS) and adipocyte (AD) differentiation in osteoporosis. A mouse model of disuse osteoporosis was made by hindlimbs unloading (HLU)/Tail - suspension. Micro - CT and histological analysis were done, and differentially expressed genes (DEGs) from GSE100930 were analyzed. Soft clustering on GSE80614 OS/AD samples found FA - related candidate genes. protein-protein interaction (PPI) network and cytoHubba's Degree algorithm identified key FA - genes, validated by quantitative polymerase chain reaction (qPCR). Key OS/AD - associated cells were identified by single - cell analysis. The mouse model showed decreased bone density, microstructure damage, increased marrow adiposity, and altered gene expression. Key FA - related genes for osteogenesis (ITGB3, LAMC1, COL6A3, ITGA8, PDGFRB) and adipogenesis (ITGB3, ITGA4, LAMB1, ITGA8, LAMA4) were found and validated. Key cells (chondrocyte, adipocyte, and osteoblast progenitors) are involved in specific pathways, with osteoblast progenitors having stronger interactions. Pseudotime analysis implies differentiation from chondrocyte progenitors to adipocyte, then osteoblast progenitors. This study provides new insights for disuse osteoporosis research.
Collapse
Affiliation(s)
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
McCallum L, Fox SW. d-limonene suppresses RANKL-induced osteoclast differentiation and promotes osteoblast activity in vitro. Biosci Biotechnol Biochem 2025; 89:232-240. [PMID: 39533827 DOI: 10.1093/bbb/zbae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Treatments for osteoporosis are typically given postfracture. Therefore, identifying safe prophylactic interventions to reduce fracture risk would be beneficial. One approach is to utilize the bioactive properties of natural compounds to modify osteoclast and osteoblast activity. d-limonene a well-tolerated, anti-inflammatory monoterpene found in citrus fruits holds promise due to its suppressive effect on NFκB, a key regulator of bone cell activity. We found that limonene promoted osteoblast differentiation and bone nodule formation and inhibited RANKL-induced osteoclast formation and bone resorption in vitro. Limonene also reduced the proresorptive signal provided by osteoblast, augmenting markers of osteoblast differentiation (alkaline phosphatase, osterix, and osteocalcin) and significantly decreasing osteoclastogenic cytokine production (PTHrP, IL-1β, and TNF-α). Therefore, limonene supplementation represents a potential route in combination with current interventions to optimize bone cell activity to maintain or enhance bone mass.
Collapse
Affiliation(s)
- Lynn McCallum
- Agri-Pharmacy Group, School of Biomedical Sciences, Faculty of Health, University of Plymouth, Plymouth, UK
| | - Simon W Fox
- Agri-Pharmacy Group, School of Biomedical Sciences, Faculty of Health, University of Plymouth, Plymouth, UK
| |
Collapse
|
3
|
Noguchi R, Osaki J, Ono T, Adachi Y, Iwata S, Yoshimatsu Y, Sasaki K, Kawai A, Kondo T. Pharmacoproteogenomic approach identifies on-target kinase inhibitors for cancer drug repositioning. In Vitro Cell Dev Biol Anim 2024; 60:1200-1214. [PMID: 39422823 DOI: 10.1007/s11626-024-00983-3] [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: 07/02/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
Drug repositioning of approved drugs offers advantages over de novo drug development for a rare type of cancer. To efficiently identify on-target drugs from clinically successful kinase inhibitors in cancer drug repositioning, drug screening and molecular profiling of cell lines are essential to exclude off-targets. We developed a pharmacoproteogenomic approach to identify on-target kinase inhibitors, combining molecular profiling of genomic features and kinase activity, and drug screening of patient-derived cell lines. This study examined eight patient-derived giant cell tumor of the bone (GCTB) cell lines, all of which harbored a signature mutation of H3-3A but otherwise without recurrent copy number variants and mutations. Kinase activity profiles of 100 tyrosine kinases with a three-dimensional substrate peptide array revealed that nine kinases were highly activated. Pharmacological screening of 60 clinically used kinase inhibitors found that nine drugs directed at 29 kinases strongly suppressed cell viability. We regarded ABL1, EGFR, and LCK as on-target kinases; among the two corresponding on-target kinase inhibitors, osimertinib and ponatinib emerged as on-target drugs whose target kinases were significantly activated. The remaining 26 kinases and seven kinase inhibitors were excluded as off-targets. Our pharmacoproteomic approach enabled the identification of on-target kinase inhibitors that are useful for drug repositioning.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Yoshimatsu
- Department of Patient-Derived Cancer Model, Tochigi Cancer Center, 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Kazuki Sasaki
- Department of Oncopeptidomics, Tochigi Cancer Center; 4-9-13 Yohnan, Utsunomiya, Tochigi, 320-0834, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
4
|
Williams KS, Seawell JA, Zhuravleva V, Pierre K, Meeker RB. Cooperative interactions between neurotrophin receptors and CXCR4 regulate macrophage phenotype and susceptibility to activation by HIV. J Neurovirol 2024; 30:406-422. [PMID: 38822196 DOI: 10.1007/s13365-024-01211-0] [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: 01/02/2024] [Revised: 04/24/2024] [Accepted: 05/08/2024] [Indexed: 06/02/2024]
Abstract
Neural damage due to inflammatory activation of macrophages and microglia is a consequence of HIV infection that leads to cognitive dysfunction. The damage is due, in part, to the release of factors that impair neuronal function but the mechanisms that control their release are poorly understood. Previous studies have shown that mature nerve growth factor (NGF) binding to tropomyosin receptor kinase A (TrkA), and proNGF acting through the p75 neurotrophin receptor (p75NTR) differentially control the phenotype of macrophages in response to HIV. However, the mechanisms responsible for these actions are unclear. The current studies demonstrated that in human monocyte-derived macrophages, CCR5 tropic HIV virions interact with the CXCR4 receptor to promote a neurotoxic macrophage phenotype. TrkA cooperatively interacted with CXCR4 to promote quick and dynamic changes in CXCR4 phosphorylation and more stable downstream actin remodeling in the form of membrane ruffles. TrkA signaling also promoted increased moacrophage calcium spiking, and low neurotoxic activity. Disruption of these interactions by HIV led to an alternative podosome-bearing phenotype with minimal calcium signaling and enhanced toxicity. Neurotrophin receptors provide an independent yet cooperative pathway for modifying the actin cytoskeleton in response to chemokines and subsequent degenerative activity. The strong opposing effects of mature and proneurotrophins may provide the opportunity to develop novel therapies that regulate the phenotype of macrophages in the context of HIV infection and perhaps other degenerative diseases.
Collapse
Affiliation(s)
- Kimberly S Williams
- Environmental and Health Sciences Program, Spelman College, 30314, Atlanta, GA, USA.
| | - Jaimie A Seawell
- Department of Neurology, University of North Carolina, 27599, Chapel Hill, NC, USA
- School of Medicine, Now at University of South Carolina, 29203, Columbia, SC, USA
| | - Viktoriya Zhuravleva
- Department of Neurology, University of North Carolina, 27599, Chapel Hill, NC, USA
- Columbia University, 10027, New York, NY, USA
| | - Kersten Pierre
- Environmental and Health Sciences Program, Spelman College, 30314, Atlanta, GA, USA
- Emory University, 30322, Atlanta, GA, USA
| | - Rick B Meeker
- Department of Neurology, University of North Carolina, 27599, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Liu Z, Ruan Z, Long H, Zhao R, Zhu Y, Lin Z, Chen P, Zhao S. Identification of ceRNA networks in type H and L vascular endothelial cells through integrated bioinformatics methods. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:562-577. [PMID: 39019785 PMCID: PMC11255190 DOI: 10.11817/j.issn.1672-7347.2024.230343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Indexed: 07/19/2024]
Abstract
OBJECTIVES Type H blood vessels are a subtype of bone-specific microvessels (CD31hiEmcnhi) that play an important regulatory role in the coupling of angiogenesis and osteogenesis. Despite reports on the distinct roles of type H and L vessels under physiological and pathological bone conditions, their genetic differences remain to be elucidated. This study aims to construct a competitive endogenous RNA (ceRNA) network of key gene for differencial expression (DE) in type H and L vascular endothelial cells (ECs) through integrated bioinformatic methods. METHODS We downloaded relevant raw data from the ArrayExpress and the Gene Expression Omnibus (GEO) database and used the Limma R-Bioconductor package to screen for DE lncRNAs, DE miRNAs, and DE mRNAs between type H and L vascular ECs. A total ceRNA network was constructed based on their interactions, followed by refinement using protein-protein interaction (PPI) networks to select upregulated and downregulated key genes. Enrichment analysis was performed on these key genes. Random validation was conducted using flow cytometry and real-time RT-PCR. RESULTS A total of 1 761 DE mRNAs, 187 DE lncRNAs, and 159 DE miRNAs were identified, and a comprehensive ceRNA network was constructed based on their interactions. Six upregulated (Itga5, Kdr, Tjp1, Pecam1, Cdh5, and Ptk2) and 2 downregulated (Csf1r and Il10) key genes were selected via PPI network to construct a subnetwork of ceRNAs related to these key genes. Upregulated key genes were mainly enriched in negative regulation of angiogenesis and vascular apoptosis. Results from flow cytometry and real-time RT-PCR were consistent with bioinformatics analysis. CONCLUSIONS This study proposes a ceRNA network associated with upregulated and downregulated type H and L vascular ECs based on selected key genes, providing new insights into the regulatory mechanisms of type H and L vascular ECs in bone metabolism.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008.
| | - Zhe Ruan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008.
| | - Haitao Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008
| | - Ruibo Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008
| | - Yong Zhu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008
| | - Zhangyuan Lin
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008
| | - Peng Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Shushan Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
6
|
Chen S, He T, Zhong Y, Chen M, Yao Q, Chen D, Shao Z, Xiao G. Roles of focal adhesion proteins in skeleton and diseases. Acta Pharm Sin B 2023; 13:998-1013. [PMID: 36970189 PMCID: PMC10031257 DOI: 10.1016/j.apsb.2022.09.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The skeletal system, which contains bones, joints, tendons, ligaments and other elements, plays a wide variety of roles in body shaping, support and movement, protection of internal organs, production of blood cells and regulation of calcium and phosphate metabolism. The prevalence of skeletal diseases and disorders, such as osteoporosis and bone fracture, osteoarthritis, rheumatoid arthritis, and intervertebral disc degeneration, increases with age, causing pain and loss of mobility and creating a huge social and economic burden globally. Focal adhesions (FAs) are macromolecular assemblies that are composed of the extracellular matrix (ECM), integrins, intracellular cytoskeleton and other proteins, including kindlin, talin, vinculin, paxillin, pinch, Src, focal adhesion kinase (FAK) and integrin-linked protein kinase (ILK) and other proteins. FA acts as a mechanical linkage connecting the ECM and cytoskeleton and plays a key role in mediating cell-environment communications and modulates important processes, such as cell attachment, spreading, migration, differentiation and mechanotransduction, in different cells in skeletal system by impacting distinct outside-in and inside-out signaling pathways. This review aims to integrate the up-to-date knowledge of the roles of FA proteins in the health and disease of skeletal system and focuses on the specific molecular mechanisms and underlying therapeutic targets for skeletal diseases.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
7
|
He Y, Cheng D, Lian C, Liu Y, Luo W, Wang Y, Ma C, Wu Q, Tian P, He D, Jia Z, Lv X, Zhang X, Pan Z, Lu J, Xiao Y, Zhang P, Liang Y, Yang Q, Hu G. Serglycin induces osteoclastogenesis and promotes tumor growth in giant cell tumor of bone. Cell Death Dis 2021; 12:868. [PMID: 34556636 PMCID: PMC8460728 DOI: 10.1038/s41419-021-04161-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/24/2021] [Accepted: 09/08/2021] [Indexed: 11/09/2022]
Abstract
Giant cell tumor of bone (GCTB) is an aggressive osteolytic bone tumor characterized by the within-tumor presence of osteoclast-like multinucleated giant cells (MGCs), which are induced by the neoplastic stromal cells and lead to extensive bone destruction. However, the underlying mechanism of the pathological process of osteoclastogenesis in GCTB is poorly understood. Here we show that the proteoglycan Serglycin (SRGN) secreted by neoplastic stromal cells plays a crucial role in the formation of MGCs and tumorigenesis in GCTB. Upregulated SRGN expression and secretion are observed in GCTB tumor cells and patients. Stromal-derived SRGN promotes osteoclast differentiation from monocytes. SRGN knockdown in stromal cells inhibits tumor growth and bone destruction in a patient-derived orthotopic xenograft model of mice. Mechanistically SRGN interacts with CD44 on the cell surface of monocytes and thus activates focal adhesion kinase (FAK), leading to osteoclast differentiation. Importantly, blocking CD44 with a neutralizing antibody reduces the number of MGCs and suppresses tumorigenesis in vivo. Overall, our data reveal a mechanism of MGC induction in GCTB and support CD44-targeting approaches for GCTB treatment.
Collapse
Affiliation(s)
- Yunfei He
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dongdong Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cheng Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingjie Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenqian Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chengxin Ma
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiuyao Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pu Tian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dasa He
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenchang Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xianzhe Lv
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xue Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Pan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jinxi Lu
- Department of General Surgery, Xinzhou District People's Hospital, Wuhan, China
| | - Yansen Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peiyuan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yajun Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingcheng Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China. .,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
8
|
Bhuyan F, de Jesus AA, Mitchell J, Leikina E, VanTries R, Herzog R, Onel KB, Oler A, Montealegre Sanchez GA, Johnson KA, Bichell L, Marrero B, De Castro LF, Huang Y, Calvo KR, Collins MT, Ganesan S, Chernomordik LV, Ferguson PJ, Goldbach-Mansky R. Novel Majeed Syndrome-Causing LPIN2 Mutations Link Bone Inflammation to Inflammatory M2 Macrophages and Accelerated Osteoclastogenesis. Arthritis Rheumatol 2021; 73:1021-1032. [PMID: 33314777 PMCID: PMC8252456 DOI: 10.1002/art.41624] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 12/10/2020] [Indexed: 12/29/2022]
Abstract
Objective To identify novel heterozygous LPIN2 mutations in a patient with Majeed syndrome and characterize the pathomechanisms that lead to the development of sterile osteomyelitis. Methods Targeted genetic analysis and functional studies assessing monocyte responses, macrophage differentiation, and osteoclastogenesis were conducted to compare the pathogenesis of Majeed syndrome to interleukin‐1 (IL‐1)–mediated diseases including neonatal‐onset multisystem inflammatory disease (NOMID) and deficiency of the IL‐1 receptor antagonist (DIRA). Results A 4‐year‐old girl of mixed ethnic background presented with sterile osteomyelitis and elevated acute‐phase reactants. She had a 17.8‐kb deletion on the maternal LPIN2 allele and a splice site mutation, p.R517H, that variably spliced out exons 10 and 11 on the paternal LPIN2 allele. The patient achieved long‐lasting remission receiving IL‐1 blockade with canakinumab. Compared to controls, monocytes and monocyte‐derived M1‐like macrophages from the patient with Majeed syndrome and those with NOMID or DIRA had elevated caspase 1 activity and IL‐1β secretion. In contrast, lipopolysaccharide‐stimulated, monocyte‐derived, M2‐like macrophages from the patient with Majeed syndrome released higher levels of osteoclastogenic mediators (IL‐8, IL‐6, tumor necrosis factor, CCL2, macrophage inflammatory protein 1α/β, CXCL8, and CXCL1) compared to NOMID patients and healthy controls. Accelerated osteoclastogenesis in the patient with Majeed syndrome was associated with higher NFATc1 levels, enhanced JNK/MAPK, and reduced Src kinase activation, and partially responded to JNK inhibition and IL‐1 (but not IL‐6) blockade. Conclusion We report 2 novel compound heterozygous disease‐causing mutations in LPIN2 in an American patient with Majeed syndrome. LPIN2 deficiency drives differentiation of proinflammatory M2‐like macrophages and enhances intrinsic osteoclastogenesis. This provides a model for the pathogenesis of sterile osteomyelitis which differentiates Majeed syndrome from other IL‐1–mediated autoinflammatory diseases.
Collapse
Affiliation(s)
- Farzana Bhuyan
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Adriana A de Jesus
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Jacob Mitchell
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Evgenia Leikina
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Rachel VanTries
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | | | - Andrew Oler
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Kim A Johnson
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Lena Bichell
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Bernadette Marrero
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Yan Huang
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Michael T Collins
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Sundar Ganesan
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Leonid V Chernomordik
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | | | | |
Collapse
|
9
|
Kreps LM, Addison CL. Targeting Intercellular Communication in the Bone Microenvironment to Prevent Disseminated Tumor Cell Escape from Dormancy and Bone Metastatic Tumor Growth. Int J Mol Sci 2021; 22:ijms22062911. [PMID: 33805598 PMCID: PMC7998601 DOI: 10.3390/ijms22062911] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
Metastasis to the bone is a common feature of many cancers including those of the breast, prostate, lung, thyroid and kidney. Once tumors metastasize to the bone, they are essentially incurable. Bone metastasis is a complex process involving not only intravasation of tumor cells from the primary tumor into circulation, but extravasation from circulation into the bone where they meet an environment that is generally suppressive of their growth. The bone microenvironment can inhibit the growth of disseminated tumor cells (DTC) by inducing dormancy of the DTC directly and later on following formation of a micrometastatic tumour mass by inhibiting metastatic processes including angiogenesis, bone remodeling and immunosuppressive cell functions. In this review we will highlight some of the mechanisms mediating DTC dormancy and the complex relationships which occur between tumor cells and bone resident cells in the bone metastatic microenvironment. These inter-cellular interactions may be important targets to consider for development of novel effective therapies for the prevention or treatment of bone metastases.
Collapse
Affiliation(s)
- Lauren M. Kreps
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Christina L. Addison
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada;
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L6, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
- Correspondence: ; Tel.: +1-613-737-7700
| |
Collapse
|
10
|
van Gastel J, Leysen H, Boddaert J, Vangenechten L, Luttrell LM, Martin B, Maudsley S. Aging-related modifications to G protein-coupled receptor signaling diversity. Pharmacol Ther 2020; 223:107793. [PMID: 33316288 DOI: 10.1016/j.pharmthera.2020.107793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Aging is a highly complex molecular process, affecting nearly all tissue systems in humans and is the highest risk factor in developing neurodegenerative disorders such as Alzheimer's and Parkinson's disease, cardiovascular disease and Type 2 diabetes mellitus. The intense complexity of the aging process creates an incentive to develop more specific drugs that attenuate or even reverse some of the features of premature aging. As our current pharmacopeia is dominated by therapeutics that target members of the G protein-coupled receptor (GPCR) superfamily it may be prudent to search for effective anti-aging therapeutics in this fertile domain. Since the first demonstration of GPCR-based β-arrestin signaling, it has become clear that an enhanced appreciation of GPCR signaling diversity may facilitate the creation of therapeutics with selective signaling activities. Such 'biased' ligand signaling profiles can be effectively investigated using both standard molecular biological techniques as well as high-dimensionality data analyses. Through a more nuanced appreciation of the quantitative nature across the multiple dimensions of signaling bias that drugs possess, researchers may be able to further refine the efficacy of GPCR modulators to impact the complex aberrations that constitute the aging process. Identifying novel effector profiles could expand the effective pharmacopeia and assist in the design of precision medicines. This review discusses potential non-G protein effectors, and specifically their potential therapeutic suitability in aging and age-related disorders.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Jan Boddaert
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, Antwerp, Belgium
| | - Laura Vangenechten
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes & Medical Genetics, Medical University of South Carolina, USA
| | - Bronwen Martin
- Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
11
|
Involvement of the FAK Network in Pathologies Related to Altered Mechanotransduction. Int J Mol Sci 2020; 21:ijms21249426. [PMID: 33322030 PMCID: PMC7764271 DOI: 10.3390/ijms21249426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023] Open
Abstract
Mechanotransduction is a physiological process in which external mechanical stimulations are perceived, interpreted, and translated by cells into biochemical signals. Mechanical stimulations exerted by extracellular matrix stiffness and cell–cell contacts are continuously applied to living cells, thus representing a key pivotal trigger for cell homeostasis, survival, and function, as well as an essential factor for proper organ development and metabolism. Indeed, a deregulation of the mechanotransduction process consequent to gene mutations or altered functions of proteins involved in perceiving cellular and extracellular mechanics can lead to a broad range of diseases, from muscular dystrophies and cardiomyopathies to cancer development and metastatization. Here, we recapitulate the involvement of focal adhesion kinase (FAK) in the cellular conditions deriving from altered mechanotransduction processes.
Collapse
|
12
|
Zhang Y, Chen XF, Li J, He F, Li X, Guo Y. lncRNA Neat1 Stimulates Osteoclastogenesis Via Sponging miR-7. J Bone Miner Res 2020; 35:1772-1781. [PMID: 32353178 DOI: 10.1002/jbmr.4039] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/10/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022]
Abstract
Increasing evidence uncover the essential role of long noncoding RNA (lncRNAs) in bone metabolism and the association of lncRNA with genetic risk of osteoporosis. However, whether lncRNA nuclear paraspeckle assembly transcript 1 (Neat1) is involved remains largely unknown. In the present study, we found that Neat1 is induced by osteoclastic differentiation stimuli. Knockdown of Neat1 attenuates osteoclast formation whereas overexpression of Neat1 accelerates osteoclast formation. In vivo evidence showed that enhanced Neat1 expression stimulates osteoclastogenesis and reduces bone mass in mice. Mechanically, Neat1 competitively binds with microRNA 7 (miR-7) and blocks its function for regulating protein tyrosine kinase 2 (PTK2). Intergenic SNP rs12789028 acts as allele-specific long-range enhancer for NEAT1 via chromatin interactions. We establish for the first time that Neat1 plays an essential role in osteoclast differentiation, and provide genetic mechanism underlying the association of NEAT1 locus with osteoporosis risk. These results enrich the current knowledge of NEAT1 function, and uncover the potential of NEAT1 as a therapeutic target for osteoporosis. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Jing Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Fang He
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
13
|
Zeng Q, Lu W, Deng Z, Wu J, Guo R, Xu X. Tablysin-15 inhibits osteoclastogenesis and LPS-induced bone loss via attenuating the integrin α vβ 3 pathway. Chem Biol Interact 2020; 327:109179. [PMID: 32534990 DOI: 10.1016/j.cbi.2020.109179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/25/2020] [Accepted: 06/09/2020] [Indexed: 01/15/2023]
Abstract
Excessive osteoclast leads to the imbalance in bone reconstruction and results in osteolytic diseases, such as osteoporosis and rheumatic arthritis. Integrin αvβ3 abundantly expresses on osteoclast and plays a critical role in the formation and function of osteoclast, therefore, blockage of αvβ3 has become an attractive therapeutic option for osteolytic diseases. In this study, we find that Tablysin-15, a RGD motif containing disintegrin, concentration-dependently suppresses RANKL-induced osteoclastogenesis, F-actin ring formation and bone resorption without affecting the cell viabilities. Tablysin-15 binds to integrin αvβ3 and inhibits the activation of FAK-associated signaling pathways. Tablysin-15 also suppresses the activation of NF-кB, MAPK, and Akt-NFATc1 signaling pathways, which are crucial transcription factors during osteoclast differentiation. Moreover, Tablysin-15 decreases the osteoclastogenesis marker gene expression, including MMP-9, TRAP, CTSK, and c-Src. Finally, Tablysin-15 significantly inhibits LPS-induced bone loss in a mouse model. Taken together, our results indicate that Tablysin-15 significantly suppresses osteoclastogenesis in vitro and in vivo, thus it might be a excellent candidate for treating osteolytic-related diseases.
Collapse
Affiliation(s)
- Qingye Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wancheng Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhenhui Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ruiyin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
14
|
Gingipains promote RANKL-induced osteoclastogenesis through the enhancement of integrin β3 in RAW264.7 cells. J Mol Histol 2020; 51:147-159. [PMID: 32193744 DOI: 10.1007/s10735-020-09865-w] [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] [Received: 01/19/2020] [Accepted: 03/10/2020] [Indexed: 01/10/2023]
Abstract
As a crucial virulence factor of Porphyromonas gingivalis, gingipains play an important role in periodontal destruction. This study aimed to investigate the effect of gingipains on osteoclastogenesis. We used RAW264.7 cells as osteoclast precursors in our study. In experimental groups, cells were treated with gingipains and/or receptor activator of nuclear factor-κB ligand (RANKL). Tartrate-resistant acid phosphatase (TRAP) activity staining assay showed osteoclast precursors and RANKL-induced mature osteoclasts were increased in a gingipains dose-dependent manner. Real-time reverse transcription polymerase chain reaction analysis demonstrated that gingipains upregulated osteoclastic genes including the protease cathepsin K (Ctsk), matrix metalloprotein 9 (Mmp9), nuclear factor of activated T cells 1 (Nfatc1) and acid phosphatase 5, tartrate resistant (Acp5) in a time-dependent manner. Western blotting assays presented upregulated expressions of TNF receptor-activating factor 6 (TRAF6) and integrin β3 induced by gingipains and RANKL compared to RANKL alone. Enhanced integrin-related signaling was also demonstrated by elevated phosphorylations of FAK and paxillin compared to control. Moreover, the pit resorption assays showed that gingipains augmented bone resorptive function of osteoclasts induced by RANKL. When we used Cilengitide to block integrin αvβ3, gingipains reversed the reduction of formation and resorptive function in RANKL-induced osteoclasts, as they enhanced integrin αvβ3 levels more than RANKL treatment alone. In conclusion, our data suggest that gingipains augmented the differentiation and function of mature osteoclasts induced by RANKL through the increase in integrin αvβ3.
Collapse
|
15
|
Choi MH, Lee K, Kim MY, Shin HI, Jeong D. Pisidium coreanum Inhibits Multinucleated Osteoclast Formation and Prevents Estrogen-Deficient Osteoporosis. Int J Mol Sci 2019; 20:ijms20236076. [PMID: 31810213 PMCID: PMC6929078 DOI: 10.3390/ijms20236076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 01/25/2023] Open
Abstract
Mollusks have served as important sources of human food and medicine for a long time. Raw Pisidium coreanum, a freshwater bivalve of the phylum Mollusca, is used in traditional therapies in parts of Asia. However, the therapeutic effects of Pisidium coreanum on bone diseases are not known. We investigated the functional roles of Pisidium coreanum in osteoporotic bone diseases. Pisidium coreanum inhibited the differentiation of bone marrow-derived monocytic cells into mature osteoclasts in vitro. The ovariectomized mice that received oral administration of Pisidium coreanum showed improvements in both trabecular and cortical bones. This preventive activity of Pisidium coreanum against bone loss was due to limited osteoclast maturation with reduced osteoclast surface extent in trabecular bone tissue. The formation of large multinucleated osteoclasts in vitro was significantly decreased in response to Pisidium coreanum, consistent with the reduced expression levels of osteoclast markers and fusion-related genes, such as NFATc1, p65, integrinαvβ3, DC-STAMP, OC-STAMP, Atp6v0d2, FAK, CD44, and MFR. These data suggest that Pisidium coreanum inhibits osteoclast differentiation by negatively regulating the fusion of mononuclear osteoclast precursors. Thus, our data demonstrate the ability of Pisidium coreanum to effectively prevent estrogen-deficient osteoporosis through inhibition of multinucleated osteoclast formation.
Collapse
Affiliation(s)
- Mun Hwan Choi
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea; (M.H.C.); (K.L.); (M.Y.K.)
| | - Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea; (M.H.C.); (K.L.); (M.Y.K.)
| | - Mi Yeong Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea; (M.H.C.); (K.L.); (M.Y.K.)
| | - Hong-In Shin
- Department of Oral Pathology, Institute for Hard Tissue and Bio-Tooth Regeneration, School of Dentistry, Kyungpook National University, Daegu 41940, Korea;
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea; (M.H.C.); (K.L.); (M.Y.K.)
- Correspondence: ; Tel.: +82-53-640-6944; Fax: +82-53-657-6869
| |
Collapse
|
16
|
Buarque de Gusmão CV, Batista NA, Vidotto Lemes VT, Maia Neto WL, de Faria LD, Alves JM, Belangero WD. Effect of Low-Intensity Pulsed Ultrasound Stimulation, Extracorporeal Shockwaves and Radial Pressure Waves on Akt, BMP-2, ERK-2, FAK and TGF-β1 During Bone Healing in Rat Tibial Defects. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2140-2161. [PMID: 31101448 DOI: 10.1016/j.ultrasmedbio.2019.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/24/2019] [Accepted: 04/07/2019] [Indexed: 06/09/2023]
Abstract
An experimental study was conducted to determine whether low-intensity pulsed ultrasound stimulation (LIPUS), extracorporeal shockwave treatment (ESWT) and radial pressure wave treatment (RPWT) modulate Akt, bone morphogenetic protein-2 (BMP-2), extracellular signal-regulated kinase-2 (ERK-2), focal adhesion kinase (FAK) and transforming growth factor-β1 (TGF-β1) during bone healing in rat tibial defects. Rat tibial defects were exposed to 500 shots of ESWT delivered at 0.12 mJ/mm2, 500 impulses of RPWT operated at 2.0 bar or to daily 20-min 30 mW/cm2 LIPUS. Following 1, 3 and 6 wk, bones were harvested to determine the expression and activity of Akt, BMP-2, ERK-2, FAK and TGF-β1. Animals exposed to ultrasound were followed up to 3 wk. Protein expression and activity were unchanged following LIPUS treatment. ESWT increased Akt activity 2.11-fold (p = 0.043) and TGF-β1 expression 9.11-fold (p = 0.016) at 1 wk and increased FAK activity 2.16-fold (p = 0.047) at 3 wk. RPWT increased FAK activity 2.6-fold (p = 0.028) at 3 wk and decreased Akt expression 0.52-fold (p = 0.05) at 6 wk. In conclusion, the protocols employed for ESWT and RPWT modulated distinct signaling pathways during fracture healing, while LIPUS standard protocol did not change the usual signaling pathways of the proteins investigated. Future studies are required to monitor osteogenesis so that the biologic meaning of our results can be clarified.
Collapse
Affiliation(s)
- Carlos Vinícius Buarque de Gusmão
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Nilza Alzira Batista
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Valeria Trombini Vidotto Lemes
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Wilson Leite Maia Neto
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Lidia Dornelas de Faria
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - José Marcos Alves
- Electrical Engineering Department, College of Engineering of São Carlos, University of São Paulo (USP), São Carlos, São Paulo, Brazil
| | - William Dias Belangero
- Department of Orthopedics and Traumatology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
17
|
Sergi C, Shen F, Liu SM. Insulin/IGF-1R, SIRT1, and FOXOs Pathways-An Intriguing Interaction Platform for Bone and Osteosarcoma. Front Endocrinol (Lausanne) 2019; 10:93. [PMID: 30881341 PMCID: PMC6405434 DOI: 10.3389/fendo.2019.00093] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/01/2019] [Indexed: 12/25/2022] Open
Abstract
Aging is a substantial risk factor for the development of osteoarthritis (OA) and, probably, an essential substrate for the development of neoplastic disease of the bone, such as osteosarcoma, which is the most common malignant mesenchymal primary bone tumor. Genetic studies have established that the insulin/insulin-like growth factor 1 (IGF-1)/phosphatidylinositol-3 kinase (PI3K)/AKT (Protein Kinase B) signal transduction pathway is involved across species, including nematodes, fruit flies, and mammals. SIRT1, a phylogenetically-conserved family of deacetylases, seems to play pleiotropic effects in epithelial malignancies of the liver and interact with the IGF-1/PI3K/AKT signal transduction pathway. Some of the most critical processes in degenerative conditions may indeed include the insulin/IGF1R and SIRT1 signaling pathways as well as some specific transcription factors. The Forkhead box O (FOXO) transcription factors (FOXOs) control diverse cellular functions, such as metabolism, longevity, and cell death. FOXOs play a critical role in the IGF-1/PI3K/AKT signal transduction pathway. FOXOs can indeed be modulated to reduce age-related diseases. FOXOs have advantageous inhibitory effects on fibroblast and myofibroblast activation, which are accompanied by a subsequent excessive production of extracellular matrix. FOXOs can block or decrease the fibrosis levels in numerous organs. Previously, we observed a correlation between nuclear FOXO3 and high caspase-8 expression, which induces cellular apoptosis in response to harmful external stimuli. In this perspective, we emphasize the current advances and interactions involving the insulin/IGF1R, SIRT1, and FOXOs pathways in the bone and osteosarcoma for a better understanding of the mechanisms potentially underpinning tissue degeneration and tumorigenesis.
Collapse
Affiliation(s)
- Consolato Sergi
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, Stollery Children's Hospital, Edmonton, AB, Canada
| | - Fan Shen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Song-Mei Liu
- Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Mori H, Hamamura K, Yo S, Hamajima K, Ootani K, Honda M, Ishizuka K, Kondo H, Tanaka K, Kodama D, Hirai T, Miyazawa K, Goto S, Togari A. Conditioned medium from rat dental pulp reduces the number of osteoclasts via attenuation of adhesiveness in osteoclast precursors. J Oral Sci 2018; 60:352-359. [PMID: 29984785 DOI: 10.2334/josnusd.17-0342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Dental pulp is known to play crucial roles in homeostasis of teeth and periodontal tissue. Although resorption of bone around the roots of nonvital teeth is occasionally observed in clinical practice, little is known about the role of dental pulp in osteoclastogenesis. Here we evaluated the effects of conditioned medium (CM) from rat dental pulp on osteoclastogenesis. It was found that the CM reduced the number of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclasts, but did not alter the mRNA levels of nuclear factor of activated T-cells, cytoplasmic 1 and TRAP. To further understand the mechanism behind these results, we evaluated the effects of CM on osteoclast precursors and found that the CM removed cell processes, resulting in a significant reduction in the number of attached cells and an increase in the number of freely floating cells. Furthermore, the CM suppressed the mRNA levels of focal adhesion kinase and paxillin, which are involved in cell adhesiveness and spreading. Collectively, the present results show that CM from dental pulp serves as an inhibitor of osteoclastogenesis by reducing the number and adhesiveness of osteoclast precursors, suggesting novel therapeutic applicability for osteoporosis.
Collapse
Affiliation(s)
- Hironori Mori
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University.,Department of Orthodontics, School of Dentistry, Aichi Gakuin University
| | - Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Shoyoku Yo
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University.,Department of Orthodontics, School of Dentistry, Aichi Gakuin University
| | - Kosuke Hamajima
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University.,Department of Orthodontics, School of Dentistry, Aichi Gakuin University
| | | | - Masaki Honda
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University
| | - Kyoko Ishizuka
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Hisataka Kondo
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Kenjiro Tanaka
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Daisuke Kodama
- Laboratory of Neuropharmacology, School of Pharmacy, Aichi Gakuin University
| | - Takao Hirai
- Laboratory of Medical Resources, School of Pharmacy, Aichi Gakuin University
| | - Ken Miyazawa
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University
| | - Shigemi Goto
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University
| | - Akifumi Togari
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| |
Collapse
|
19
|
Park OJ, Kim J, Ahn KB, Lee JY, Park YJ, Kum KY, Yun CH, Han SH. A 15-amino acid C-terminal peptide of beta-defensin-3 inhibits bone resorption by inhibiting the osteoclast differentiation and disrupting podosome belt formation. J Mol Med (Berl) 2017; 95:1315-1325. [DOI: 10.1007/s00109-017-1589-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 02/05/2023]
|
20
|
Wang H, Luo J, Carlton C, McGinnis LK, Kinsey WH. Sperm-oocyte contact induces outside-in signaling via PYK2 activation. Dev Biol 2017; 428:52-62. [PMID: 28527703 PMCID: PMC5539980 DOI: 10.1016/j.ydbio.2017.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 01/20/2023]
Abstract
Fertilization is a multi-step process that begins with plasma membrane interactions that enable sperm - oocyte binding followed by fusion of the sperm and oocyte plasma membranes. Once membrane fusion has occurred, sperm incorporation involves actin remodeling events within the oocyte cortex that allow the sperm head to penetrate the cortical actin layer and gain access to the ooplasm. Despite the significance for reproduction, the control mechanisms involved in gamete binding, fusion, and sperm incorporation are poorly understood. While it is known that proline - rich tyrosine kinase 2 (PYK2 or PTK2b) kinase activity plays an important role in fertilization, its specific function has not been addressed. The present study made use of a zona-free mouse oocyte fertilization assay to investigate the relationship between PYK2 activity and sperm - oocyte binding and fusion, as well as localized changes in actin polymerization and sperm incorporation. In this assay, the majority of bound sperm had no apparent effect on the oocyte and only a few became incorporated into the ooplasm. However, a subset of bound sperm were associated with a localized response in which PYK2 was recruited to the oocyte cortex where it frequently co-localized with a ring or disk of f-actin. The frequency of sperm-oocyte binding sites that exhibited this actin response was reduced in pyk2-/- oocytes and the pyk2-/- oocytes proved less efficient at incorporating sperm, indicating that this protein kinase may have an important role in sperm incorporation. The response of PYK2 to sperm-oocyte interaction appeared unrelated to gamete fusion since PYK2 was recruited to sperm - binding sites under conditions where sperm - oocyte fusion was prevented and since PYK2 suppression or ablation did not prevent sperm - oocyte fusion. While a direct correlation between the PYK2 response in the oocyte and the successful incorporation of individual bound sperm remains to be established, these findings suggest a model in which the oocyte is not a passive participant in fertilization, but instead responds to sperm contact by localized PYK2 signaling that promotes actin remodeling events required to physically incorporate the sperm head into the ooplasm.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jinping Luo
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA; Applied StemCell Inc., Milpitas, CA 95035, USA
| | - Carol Carlton
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lynda K McGinnis
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA 90033, USA
| | - William H Kinsey
- Department of Anatomy & Cell Biology, Univ. of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
21
|
Llewellyn RA, Thomas KS, Gutknecht MF, Bouton AH. The nonreceptor protein tyrosine kinase Pyk2 promotes the turnover of monocytes at steady state. J Leukoc Biol 2017; 102:1069-1080. [PMID: 28754799 DOI: 10.1189/jlb.1a0217-063r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/20/2017] [Accepted: 07/10/2017] [Indexed: 12/30/2022] Open
Abstract
Monocytes are short-lived myeloid cells that perform functions essential for tissue homeostasis and disease resolution. However, the cellular mechanisms controlling the maintenance and turnover of monocyte populations are largely undefined. Proline-rich tyrosine kinase 2 (Pyk2) is a nonreceptor tyrosine kinase that regulates numerous immune cell functions, but its role in monocytes is currently unknown. In this study, we sought to characterize the expression and function of Pyk2 in lineage-committed monocyte populations. Here, we report that Pyk2 protein expression is increased in the Ly6C- monocyte population. Using a Pyk2 knockout mouse model (Pyk2-/-), we show that Pyk2 regulates the relative proportion of monocyte subsets normally represented in the bone marrow (BM) at steady state. In support of this conclusion, a similar phenotype was observed in the peripheral blood and spleen. Data from reciprocal BM chimera experiments indicate that the alterations in monocyte populations exhibited by Pyk2-/- mice are due to factors intrinsic to the monocytes. Lineage-tracing of monocyte populations suggests that Pyk2 promotes apoptosis in BM monocytes, thereby acting as an important homeostatic regulator of turnover in these short-lived, innate immune cells.
Collapse
Affiliation(s)
- Ryan A Llewellyn
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Keena S Thomas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Michael F Gutknecht
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amy H Bouton
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
22
|
Meirson T, Samson AO, Gil-Henn H. An in silico high-throughput screen identifies potential selective inhibitors for the non-receptor tyrosine kinase Pyk2. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1535-1557. [PMID: 28572720 PMCID: PMC5441678 DOI: 10.2147/dddt.s136150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The non-receptor tyrosine kinase proline-rich tyrosine kinase 2 (Pyk2) is a critical mediator of signaling from cell surface growth factor and adhesion receptors to cell migration, proliferation, and survival. Emerging evidence indicates that signaling by Pyk2 regulates hematopoietic cell response, bone density, neuronal degeneration, angiogenesis, and cancer. These physiological and pathological roles of Pyk2 warrant it as a valuable therapeutic target for invasive cancers, osteoporosis, Alzheimer’s disease, and inflammatory cellular response. Despite its potential as a therapeutic target, no potent and selective inhibitor of Pyk2 is available at present. As a first step toward discovering specific potential inhibitors of Pyk2, we used an in silico high-throughput screening approach. A virtual library of six million lead-like compounds was docked against four different high-resolution Pyk2 kinase domain crystal structures and further selected for predicted potency and ligand efficiency. Ligand selectivity for Pyk2 over focal adhesion kinase (FAK) was evaluated by comparative docking of ligands and measurement of binding free energy so as to obtain 40 potential candidates. Finally, the structural flexibility of a subset of the docking complexes was evaluated by molecular dynamics simulation, followed by intermolecular interaction analysis. These compounds may be considered as promising leads for further development of highly selective Pyk2 inhibitors.
Collapse
Affiliation(s)
- Tomer Meirson
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Abraham O Samson
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| | - Hava Gil-Henn
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, Israel
| |
Collapse
|
23
|
Nakamura S, Koyama T, Izawa N, Nomura S, Fujita T, Omata Y, Minami T, Matsumoto M, Nakamura M, Fujita-Jimbo E, Momoi T, Miyamoto T, Aburatani H, Tanaka S. Negative feedback loop of bone resorption by NFATc1-dependent induction of Cadm1. PLoS One 2017; 12:e0175632. [PMID: 28414795 PMCID: PMC5393607 DOI: 10.1371/journal.pone.0175632] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/29/2017] [Indexed: 11/29/2022] Open
Abstract
Trimethylation of histone H3 lysine 4 and lysine 27 (H3K4me3 and H3K27me3) at gene promoter regions critically regulates gene expression. Key developmental genes tend to exhibit changes in histone modification patterns from the H3K4me3/H3K27me3 bivalent pattern to the H3K4me3 monovalent pattern. Using comprehensive chromatin immunoprecipitation followed by sequencing in bone marrow-derived macrophages (BMMs) and mature osteoclasts, we found that cell surface adhesion molecule 1 (Cadm1) is a direct target of nuclear factor of activated T cells 1 (NFATc1) and exhibits a bivalent histone pattern in BMMs and a monovalent pattern in osteoclasts. Cadm1 expression was upregulated in BMMs by receptor activator of nuclear factor kappa B ligand (RANKL), and blocked by a calcineurin/NFATc1 inhibitor, FK506. Cadm1-deficient mice exhibited significantly reduced bone mass compared with wild-type mice, which was due to the increased osteoclast differentiation, survival and bone-resorbing activity in Cadm1-deficient osteoclasts. These results suggest that Cadm1 is a direct target of NFATc1, which is induced by RANKL through epigenetic modification, and regulates osteoclastic bone resorption in a negative feedback manner.
Collapse
Affiliation(s)
- Shinya Nakamura
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takuma Koyama
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Naohiro Izawa
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Seitaro Nomura
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Takanori Fujita
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Yasunori Omata
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Minami
- Division of Phenotype Disease Analysis, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Kumamoto, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University, Tokyo, Japan
| | - Eriko Fujita-Jimbo
- Department of Pediatrics, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Takashi Momoi
- Department of Pathophysiology, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Takeshi Miyamoto
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
24
|
Guo Y, Wang Y, Liu Y, Wang H, Guo C, Zhang X, Bei C. Effect of the same mechanical loading on osteogenesis and osteoclastogenesis in vitro. Chin J Traumatol 2017; 18:150-6. [PMID: 26643241 DOI: 10.1016/j.cjtee.2014.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE To investigate the influence of the same mechanical loading on osteogenesis and osteoclastogenesis in vitro. METHODS Primary osteoblasts, bone marrow-derived mesenchymal stem cells (BMSCs, cultured in osteoinductive medium) and RAW264.7 cells cultured in osteoclast inductive medium were all subjected to a 1000 μstrain (μs) at 1 Hz cyclic mechanical stretch for 30 min (twice a day). RESULTS After mechanical stimulation, the alkaline phosphatase (ALP) activity, osteocalcin protein level of the osteoblasts and BMSCs were all enhanced, and the mRNA levels of ALP and collagen type I increased. Additionally, extracellular-deposited calcium of both osteoblasts and BMSCs increased. At the same time, the activity of secreted tartrate-resistant acid phosphatase, the number of tartrate-resistant acid phosphatase-positive multinucleated cells, matrix metalloproteinase-9 protein levels of RAW264.7 cells and the extracellular calcium solvency all decreased. CONCLUSION The results demonstrated that 1000 μs cyclic mechanical loading enhanced osteoblasts activity, promoted osteoblastic differentiation of BMSCs and restrained osteoclastogenesis of RAW264.7 cells in vitro.
Collapse
Affiliation(s)
- Yong Guo
- College of Biotechnology, Guilin Medical University, Guilin 541004, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Wang W, Hao J, Zheng S, Fan Q, He A, Wen Y, Guo X, Wu C, Wang S, Yang T, Shen H, Chen X, Tian Q, Tan L, Deng HW, Zhang F. Tissue-specific pathway association analysis using genome-wide association study summaries. Bioinformatics 2016; 33:243-247. [PMID: 27651483 DOI: 10.1093/bioinformatics/btw595] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/20/2016] [Accepted: 09/08/2016] [Indexed: 01/25/2023] Open
Abstract
MOTIVATION Pathway association analysis has made great achievements in elucidating the genetic basis of human complex diseases. However, current pathway association analysis approaches fail to consider tissue-specificity. RESULTS We developed a tissue-specific pathway interaction enrichment analysis algorithm (TPIEA). TPIEA was applied to two large Caucasian and Chinese genome-wide association study summary datasets of bone mineral density (BMD). TPIEA identified several significant pathways for BMD [false discovery rate (FDR) < 0.05], such as KEGG FOCAL ADHESION and KEGG AXON GUIDANCE, which had been demonstrated to be involved in the development of osteoporosis. We also compared the performance of TPIEA and classical pathway enrichment analysis, and TPIEA presented improved performance in recognizing disease relevant pathways. TPIEA may help to fill the gap of classic pathway association analysis approaches by considering tissue specificity. AVAILABILITY AND IMPLEMENTATION The online web tool of TPIEA is available at https://sourceforge.net/projects/tpieav1/files CONTACT: fzhxjtu@mail.xjtu.edu.cnSupplementary information: Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Wenyu Wang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Jingcan Hao
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Shuyu Zheng
- Department of Radiation Oncology, First Affiliated Hospital, Health Science Center
| | - Qianrui Fan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Awen He
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Xiong Guo
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Cuiyan Wu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Sen Wang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| | - Tielin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Hui Shen
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine.,Center for Bioinformatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Xiangding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, P. R. China
| | - Qing Tian
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine.,Center for Bioinformatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Lijun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, P. R. China
| | - Hong-Wen Deng
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine.,Center for Bioinformatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center
| |
Collapse
|
26
|
Chiu YH, Ritchlin CT. DC-STAMP: A Key Regulator in Osteoclast Differentiation. J Cell Physiol 2016; 231:2402-7. [PMID: 27018136 DOI: 10.1002/jcp.25389] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 01/09/2023]
Abstract
Osteoimmunology research is a new emerging research field that investigates the links between the bone and immune responses. Results from osteoimmunology studies suggest that bone is not only an essential component of the musculoskeletal system, but is also actively involved in immune regulation. Many important factors involved in immune regulation also participate in bone homeostasis. Bone homeostasis is achieved by a coordinated action between bone-synthesizing osteoblasts and bone-degrading osteoclasts. An imbalanced action between osteoblasts and osteoclasts often results in pathological bone diseases: osteoporosis is caused by an excessive osteoclast activity, whereas osteopetrosis results from an increased osteoblast activity. This review focuses on dendritic cell-specific transmembrane protein (DC-STAMP), an important protein currently considered as a master regulator of osteoclastogenesis. Of clinical relevance, the frequency of circulating DC-STAMP+ cells is elevated during the pathogenesis of psoriatic diseases. Intriguingly, recent results suggest that DC-STAMP also plays an imperative role in bone homeostasis by regulating the differentiation of both osteoclasts and osteoblasts. This article summarizes our current knowledge on DC-STAMP by focusing on its interacting proteins, its regulation on osteoclastogenesis-related genes, its possible involvement in immunoreceptor tyrosine-based inhibitory motif (ITIM)-mediated signaling cascade, and its potential of developing therapeutics for clinical applications. J. Cell. Physiol. 231: 2402-2407, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ya-Hui Chiu
- Division of Allergy, Immunology and Rheumatology, School of Medicine, University of Rochester, Rochester, New York
| | - Christopher T Ritchlin
- Division of Allergy, Immunology and Rheumatology, School of Medicine, University of Rochester, Rochester, New York
| |
Collapse
|
27
|
|
28
|
Noncanonical G-protein-dependent modulation of osteoclast differentiation and bone resorption mediated by Pasteurella multocida toxin. mBio 2014; 5:e02190. [PMID: 25389180 PMCID: PMC4235216 DOI: 10.1128/mbio.02190-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pasteurella multocida toxin (PMT) induces atrophic rhinitis in animals, which is characterized by a degradation of nasal turbinate bones, indicating an effect of the toxin on bone cells such as osteoblasts and osteoclasts. The underlying molecular mechanism of PMT was defined as a persistent activation of heterotrimeric G proteins by deamidation of a specific glutamine residue. Here, we show that PMT acts directly on osteoclast precursor cells such as bone marrow-derived CD14+ monocytes and RAW246.7 cells to induce osteoclastogenesis as measured by expression of osteoclast-specific markers such as tartrate-resistant acid phosphatase and bone resorption activity. Treatment performed solely with PMT stimulates osteoclast differentiation, showing a receptor activator of nuclear factor-κB ligand (RANKL)-independent action of the toxin. The underlying signal transduction pathway was defined as activation of the heterotrimeric G proteins Gαq/11 leading to the transactivation of Ras and the mitogen-activated protein kinase pathway. Gαq/11 transactivates Ras via its effector phospholipase Cβ-protein kinase C (PKC) involving proline-rich tyrosine kinase 2 (Pyk2). PMT-induced activation of the mitogen-activated protein kinase pathway results in stimulation of the osteoclastogenic transcription factors AP-1, NF-κB, and NFATc1. In addition, Ca2+-dependent calcineurin activation of NFAT is crucial for PMT-induced osteoclastogenesis. The data not only elucidate a rationale for PMT-dependent bone loss during atrophic rhinitis but also highlight a noncanonical, G-protein-dependent pathway toward bone resorption that is distinct from the RANKL-RANK pathway but mimics it. We define heterotrimeric G proteins as as-yet-underestimated entities/players in the maturation of osteoclasts which might be of pharmacological relevance. Pasteurella multocida toxin (PMT) induces degradation of nasal turbinate bones, leading to the syndrome of atrophic rhinitis. Recently, the molecular mechanism and substrate specificity of PMT were identified. The toxin activates heterotrimeric G proteins by a covalent modification. However, the mechanism by which PMT induces bone degradation is poorly understood. Our report demonstrates a direct effect of PMT on osteoclast precursor cells, leading to maturation of bone-degrading osteoclasts. Interestingly, PMT stimulates osteoclastogenesis independently of the cytokine RANKL, which is a key factor in induction of osteoclast differentiation. This implicates a noncanonical osteoclastogenic signaling pathway induced by PMT. The elucidated Gαq/11-dependent osteoclastogenic signal transduction pathway ends in osteoclastogenic NFAT signaling. The noncanonical, heterotrimeric G protein-dependent osteoclast differentiation process may be of pharmacological relevance, as members of this pathway are highly druggable. In particular, modulation of G protein-coupled receptor activity in osteoclast progenitors by small molecules might be of specific interest.
Collapse
|
29
|
Shelef MA, Bennin DA, Yasmin N, Warner TF, Ludwig T, Beggs HE, Huttenlocher A. Focal adhesion kinase is required for synovial fibroblast invasion, but not murine inflammatory arthritis. Arthritis Res Ther 2014; 16:464. [PMID: 25280866 PMCID: PMC4203874 DOI: 10.1186/s13075-014-0464-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 09/24/2014] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Synovial fibroblasts invade cartilage and bone, leading to joint destruction in rheumatoid arthritis. However, the mechanisms that regulate synovial fibroblast invasion are not well understood. Focal adhesion kinase (FAK) has been implicated in cellular invasion in several cell types, and FAK inhibitors are in clinical trials for cancer treatment. Little is known about the role of FAK in inflammatory arthritis, but, given its expression in synovial tissue, its known role in invasion in other cells and the potential clinical availability of FAK inhibitors, it is important to determine if FAK contributes to synovial fibroblast invasion and inflammatory arthritis. METHODS After treatment with FAK inhibitors, invasiveness of human rheumatoid synovial fibroblasts was determined with Matrigel invasion chambers. Migration and focal matrix degradation, two components of cellular invasion, were assessed in FAK-inhibited rheumatoid synovial fibroblasts by transwell assay and microscopic examination of fluorescent gelatin degradation, respectively. Using mice with tumor necrosis factor α (TNFα)-induced arthritis in which fak could be inducibly deleted, invasion and migration by FAK-deficient murine arthritic synovial fibroblasts were determined as described above and arthritis was clinically and pathologically scored in FAK-deficient mice. RESULTS Inhibition of FAK in human rheumatoid synovial fibroblasts impaired cellular invasion and migration. Focal matrix degradation occurred both centrally and at focal adhesions, the latter being a novel site for matrix degradation in synovial fibroblasts, but degradation was unaltered with FAK inhibitors. Loss of FAK reduced invasion in murine arthritic synovial fibroblasts, but not migration or TNFα-induced arthritis severity and joint erosions. CONCLUSIONS FAK inhibitors reduce synovial fibroblast invasion and migration, but synovial fibroblast migration and TNFα-induced arthritis do not rely on FAK itself. Thus, inhibition of FAK alone is unlikely to be sufficient to treat inflammatory arthritis, but current drugs that inhibit FAK may inhibit multiple factors, which could increase their efficacy in rheumatoid arthritis.
Collapse
Affiliation(s)
- Miriam A Shelef
- />Division of Rheumatology, Department of Medicine, University of Wisconsin–Madison and William S Middleton Memorial VA Medical Center, 1685 Highland Ave, Madison, WI 53705 USA
| | - David A Bennin
- />Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, 1550 Linden Drive, Madison, WI 53706 USA
| | - Nihad Yasmin
- />Department of Internal Medicine, Aurora Healthcare, 945 N 12th street, Milwaukee, WI 53233 USA
| | - Thomas F Warner
- />Department of Pathology and Laboratory Medicine, University of Wisconsin – Madison, 600 Highland Ave, Madison, WI 53792 USA
| | - Thomas Ludwig
- />Department of Molecular and Cellular Biochemistry, The Ohio State University, 460 West 12th Avenue, Columbus, OH 43210 USA
| | - Hilary E Beggs
- />Department of Ophthalmology, University of California, 10 Koret Way, San Francisco, CA 94143 USA
| | - Anna Huttenlocher
- />Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, 1550 Linden Drive, Madison, WI 53706 USA
| |
Collapse
|
30
|
Mediero A, Perez-Aso M, Cronstein BN. Activation of EPAC1/2 is essential for osteoclast formation by modulating NFκB nuclear translocation and actin cytoskeleton rearrangements. FASEB J 2014; 28:4901-13. [PMID: 25122553 DOI: 10.1096/fj.14-255703] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bisphosphonates inhibit osteoclast differentiation/function via inhibition of Rap1A isoprenylation. As Rap1 is the effector of exchange protein directly activated by cAMP (EPAC) proteins, we determined the role of EPAC in osteoclast differentiation. We examined osteoclast differentiation as the number of primary murine/human bone-marrow precursors that differentiated into multinucleated TRAP-positive cells in the presence of EPAC-selective stimulus (8-pCTP-2'-O-Me-cAMP, 100 μM; 8-pCTP-2'-O-Me-cAMP-AM, 1 μM) or inhibitor brefeldin A (BFA), ESI-05, and ESI-09 (10 μM each). Rap1 activity was assessed, and signaling events, as well as differentiation in EPAC1/2-knockdown RAW264.7 cells, were studied. Direct EPAC1/2 stimulation significantly increased osteoclast differentiation, whereas EPAC1/2 inhibition diminished differentiation (113 ± 6%, P < 0.05, and 42 ± 10%, P < 0.001, of basal, respectively). Rap1 activation was maximal 15 min after RANKL stimulation (147 ± 9% of basal, P < 0.001), whereas silencing of EPAC1/2 diminished activated Rap1 (43 ± 13 and 20 ± 15% of control, P < 0.001) and NFkB nuclear translocation. TRAP-staining revealed no osteoclast differentiation in EPAC1/2-KO cells. Cathepsin K, NFATc1, and osteopontin mRNA expression decreased in EPAC1/2-KO cells when compared to control. RhoA, cdc42, Rac1, and FAK were activated in an EPAC1/2-dependent manner, and there was diminished cytoskeletal assembly in EPAC1/2-KO cells. In summary, EPAC1 and EPAC2 are critical signaling intermediates in osteoclast differentiation that permit RANKL-stimulated NFkB nuclear translocation and actin rearrangements. Targeting this signaling intermediate may diminish bone destruction in inflammatory arthritis.
Collapse
Affiliation(s)
- Aránzazu Mediero
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine, New York, New York, USA
| | - Miguel Perez-Aso
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine, New York, New York, USA
| | - Bruce N Cronstein
- Department of Medicine, Division of Translational Medicine, New York University School of Medicine, New York, New York, USA
| |
Collapse
|