1
|
Iwanowska M, Kochman M, Szatko A, Zgliczyński W, Glinicki P. Bone Disease in Primary Hyperparathyroidism-Changes Occurring in Bone Metabolism and New Potential Treatment Strategies. Int J Mol Sci 2024; 25:11639. [PMID: 39519190 PMCID: PMC11546563 DOI: 10.3390/ijms252111639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Primary hyperparathyroidism (PHPT) is a common endocrinopathy, predominantly caused by a single parathyroid adenoma that is responsible for the excessive secretion of parathyroid hormone (PTH)-the hallmark of disease. Excess of this hormone causes remarkable changes in bone metabolism, including an increased level of bone remodeling with a predominance of bone resorption. Those changes lead to deterioration of bone structure and density, especially in cortical bone. The main treatment for PHPT is surgical removal of the adenoma, which normalizes PTH levels and terminates the progression of bone disease and leads to its regeneration. However, because not all the patients are suitable candidates for surgery, alternative therapies are needed. Current non-surgical treatments targeting bone disease secondary to PHPT include bisphosphonates and denosumab. Those antiresorptives prevent further bone loss, but they lack the ability to regenerate already degraded bone. There is ongoing research to find targeted drugs capable of halting resorption alongside stimulating bone formation. This review presents the advancements in understanding the molecular mechanisms responsible for bone disease in PHPT and assesses the efficacy of new potential therapeutic approaches (e.g., allosteric inhibitors of the PTH receptor, V-ATPase, or cathepsin inhibitors) aimed at mitigating bone loss and enhancing bone regeneration in affected patients.
Collapse
Affiliation(s)
- Mirella Iwanowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Magdalena Kochman
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Alicja Szatko
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- EndoLab Laboratory, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Wojciech Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Piotr Glinicki
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- EndoLab Laboratory, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| |
Collapse
|
2
|
Xu M, Zhao J, Zhu L, Ge C, Sun Y, Wang R, Li Y, Dai X, Kuang Q, Hu L, Luo J, Kuang G, Ren Y, Wang B, Tan J, Shi S. Targeting PYK2 with heterobifunctional T6BP helps mitigate MASLD and MASH-HCC progression. J Hepatol 2024:S0168-8278(24)02538-8. [PMID: 39260704 DOI: 10.1016/j.jhep.2024.08.029] [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: 07/07/2023] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND & AIMS The mechanisms underlying the regulation of hepatocyte non-receptor tyrosine kinases in metabolic dysfunction-associated steatohepatitis (MASH) remain largely unclear. METHODS Hepatocyte-specific overexpression or deletion and anti-protein tyrosine kinase 2 beta (PYK2) or anti-TRAF6-binding protein (T6BP) crosslinking were utilised to study fatty liver protection by T6BP. P-PTC, a peptide-proteolysis targeting chimaera, degrades PYK2 to block MASH progression. RESULTS Since PYK2 activation is promoter signalling in steatohepatitis development, we find that T6BP is a novel and critical suppressor of PYK2 that reduces hepatic lipid accumulation, pro-inflammatory factor release, and pro-fibrosis production by ubiquitin ligase CBL to degrade PYK2. Mechanistic evidence suggests that T6BP directly targets PYK2 and prevents its N-terminal FERM domain-triggered dimerization, disrupting downstream PYK2-JNK signalling hyperactivation. Additionally, T6BP favourably recruits CBL, a particular E3 ubiquitin ligase targeting PYK2, to form a complex and degrade PYK2. T6BP (F1), a core fragment of T6BP, directly blocks N-terminal FERM domain-associated dimerization of PYK2, followed by T6BP-recruiting CBL-mediated PYK2 degradation in a typical T6BP-dependent manner when the tiny fragment is specifically expressed using thyroxine binding globulin (TBG)-ground vectors. This inhibits the progression of MASH, metabolic dysfunction-associated steatotic liver disease (MASLD)-related HCC (MASH-HCC), and metabolic syndrome in dietary rodent models. First-ever peptide-proteolysis targeting chimaera (P-PTC) based on the core segment of T6BP as a ligand for targeted recruitment of CBL targeting metabolic disorders like MASH has been devised and validated in animal models. CONCLUSIONS Our study revealed a previously unknown mechanism: identification of T6BP as a key eliminator of fatty liver strongly contributes to the development of promising therapeutic targets, and the discovery of crucial fragments of T6BP-based pharmacon that interrupt PYK2 dimerization are novel and viable treatments for fatty liver and its advanced symptoms and complications. IMPACT AND IMPLICATIONS Excessive high-energy diet ingestion is critical in driving steatohepatitis via regulation of hepatocyte non-receptor tyrosine kinases. The mechanisms under lying the regulation of hepatocyte PYK2 in metabolic dysfunction-associated steatohepatitis (MASH) remain largely unclear. Here, we found that T6BP as a critical fatty liver eliminator has a significant impact on the development of promising therapeutic targets. Additionally, vital T6BP-based pharmacon fragments that impede PYK2 dimerization have been found, offering new and effective treatments for advanced fatty liver symptoms and complications.
Collapse
Affiliation(s)
- Minxuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China.
| | - Junjie Zhao
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Liancai Zhu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Chenxu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Yan Sun
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Ranran Wang
- College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China; School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Collaborative Innovation Center for Child Nutrition and Health Development, Chongqing University of Education, Chongqing 400067, PR China
| | - Yuanyuan Li
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China
| | - Xianling Dai
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Qin Kuang
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Linfeng Hu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China
| | - Jing Luo
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China
| | - Gang Kuang
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China
| | - Yanrong Ren
- College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China; School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, PR China.
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; College of Modern Health Industry, Chongqing University of Education, Chongqing 400067, PR China.
| | - Shengbin Shi
- Department of Gastrointestinal Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Science, Jinan 250117, PR China.
| |
Collapse
|
3
|
Liu Y, Chen H, Chen T, Qiu G, Han Y. The emerging role of osteoclasts in the treatment of bone metastases: rationale and recent clinical evidence. Front Oncol 2024; 14:1445025. [PMID: 39148909 PMCID: PMC11324560 DOI: 10.3389/fonc.2024.1445025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
The occurrence of bone metastasis is a grave medical concern that substantially impacts the quality of life in patients with cancer. The precise mechanisms underlying bone metastasis remain unclear despite extensive research efforts, and efficacious therapeutic interventions are currently lacking. The ability of osteoclasts to degrade the bone matrix makes them a crucial factor in the development of bone metastasis. Osteoclasts are implicated in several aspects of bone metastasis, encompassing the formation of premetastatic microenvironment, suppression of the immune system, and reactivation of quiescent tumor cells. Contemporary clinical interventions targeting osteoclasts have proven effective in mitigating bone-related symptoms in patients with cancer. This review comprehensively analyzes the mechanistic involvement of osteoclasts in bone metastasis, delineates potential therapeutic targets associated with osteoclasts, and explores clinical evidence regarding interventions targeting osteoclasts.
Collapse
Affiliation(s)
- Youjun Liu
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Huanshi Chen
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Tong Chen
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Guowen Qiu
- Department of Spinal Surgery, Liuzhou Municipal Liutie Central Hospital, Liuzhou, China
| | - Yu Han
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Li Z, Yang X, Fu R, Wu Z, Xu S, Jiao J, Qian M, Zhang L, Wu C, Xie T, Yao J, Wu Z, Li W, Ma G, You Y, Chen Y, Zhang HK, Cheng Y, Tang X, Wu P, Lian G, Wei H, Zhao J, Xu J, Ai L, Siwko S, Wang Y, Ding J, Song G, Luo J, Liu M, Xiao J. Kisspeptin-10 binding to Gpr54 in osteoclasts prevents bone loss by activating Dusp18-mediated dephosphorylation of Src. Nat Commun 2024; 15:1300. [PMID: 38346942 PMCID: PMC10861593 DOI: 10.1038/s41467-024-44852-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2024] [Indexed: 02/15/2024] Open
Abstract
Osteoclasts are over-activated as we age, which results in bone loss. Src deficiency in mice leads to severe osteopetrosis due to a functional defect in osteoclasts, indicating that Src function is essential in osteoclasts. G-protein-coupled receptors (GPCRs) are the targets for ∼35% of approved drugs but it is still unclear how GPCRs regulate Src kinase activity. Here, we reveal that GPR54 activation by its natural ligand Kisspeptin-10 (Kp-10) causes Dusp18 to dephosphorylate Src at Tyr 416. Mechanistically, Gpr54 recruits both active Src and the Dusp18 phosphatase at its proline/arginine-rich motif in its C terminus. We show that Kp-10 binding to Gpr54 leads to the up-regulation of Dusp18. Kiss1, Gpr54 and Dusp18 knockout mice all exhibit osteoclast hyperactivation and bone loss, and Kp-10 abrogated bone loss by suppressing osteoclast activity in vivo. Therefore, Kp-10/Gpr54 is a promising therapeutic target to abrogate bone resorption by Dusp18-mediated Src dephosphorylation.
Collapse
Affiliation(s)
- Zhenxi Li
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| | - Xinghai Yang
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Ruifeng Fu
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Zhipeng Wu
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Shengzhao Xu
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jian Jiao
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Ming Qian
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Long Zhang
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Chunbiao Wu
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Tianying Xie
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jiqiang Yao
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Zhixiang Wu
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Wenjun Li
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guoli Ma
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yu You
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yihua Chen
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Han-Kun Zhang
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yiyun Cheng
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiaolong Tang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Pengfei Wu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Gewei Lian
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Haifeng Wei
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jian Zhao
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lianzhong Ai
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Stefan Siwko
- Department of Translational Medical Sciences, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Yue Wang
- Shanghai Key Lab of Cell Engineering; Translational Medicine Research Center, Naval Medical University, Shanghai, 200433, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Gaojie Song
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China.
| | - Mingyao Liu
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jianru Xiao
- Institute of Orthopedic Biomedical and Device Innovation, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
- Institute of Orthopedics, Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
5
|
Tang L, Li B, Su Q, Chen X, He R. Identification of hub genes and therapeutic drugs in osteonecrosis of the femoral head through integrated bioinformatics analysis and literature mining. Sci Rep 2023; 13:11972. [PMID: 37488209 PMCID: PMC10366127 DOI: 10.1038/s41598-023-39258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/22/2023] [Indexed: 07/26/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a multifactorial disease leading to severely limited function. By far, the etiology and pathogenesis of ONFH are not fully understood, and surgery is the only effective way to treat ONFH. This study aims to identify hub genes and therapeutic drugs in ONFH. Two gene expression profiles were downloaded from the gene expression omnibus database, and the hub genes and candidate drugs for ONFH were identified through integrated bioinformatics analysis and cross-validated by literature mining. A total of 159 DEGs were identified. PTGS2, LRRK2, ANXA5, IGF1R, MCL1, TIMP2, LYN, CD68, CBL, and RUNX2 were validated as 10 hub genes, which has considerable implications for future genetic research and related research fields of ONFH. Our findings indicate that 85 drugs interact with ONFH, with most drugs exhibiting a positive impact on ONFH by promoting osteogenesis and angiogenesis or inhibiting microcirculation embolism, rather than being anti-inflammatory. Our study provides novel insights into the pathogenesis, prevention, and treatment of ONFH.
Collapse
Affiliation(s)
- Lan Tang
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Bin Li
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Qiuming Su
- Department of Hepatopancreatobiliary Surgery, The First People's Hospital of Kunming, Calmette Hospital, Kunming City, Yunnan Province, China
| | - Xi Chen
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China
- Department of Epidemiology and Statistics, School of Public Health, Medical College, Zhejiang University, Hangzhou City, Zhejiang Province, China
| | - Rongxin He
- Department of Orthopedic, The Second Affiliated Hospital, Zhejiang University School of Medicine, #88 Jiefang Road, Hangzhou City, 310001, Zhejiang Province, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
6
|
Choi EB, Agidigbi TS, Kang IS, Kim C. ERK Inhibition Increases RANKL-Induced Osteoclast Differentiation in RAW 264.7 Cells by Stimulating AMPK Activation and RANK Expression and Inhibiting Anti-Osteoclastogenic Factor Expression. Int J Mol Sci 2022; 23:13512. [PMID: 36362318 PMCID: PMC9656104 DOI: 10.3390/ijms232113512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 08/13/2023] Open
Abstract
Bone absorption is necessary for the maintenance of bone homeostasis. An osteoclast (OC) is a monocyte-macrophage lineage cell that absorbs bone tissue. Extracellular signal-regulated kinases (ERKs) are known to play important roles in regulating OC growth and differentiation. In this study, we examined specific downstream signal pathways affected by ERK inhibition during OC differentiation. Our results showed that the ERK inhibitors PD98059 and U0126 increased receptor activator of NF-κB ligand (RANKL)-induced OC differentiation in RAW 264.7 cells, implying a negative role in OC differentiation. This is supported by the effect of ERK2-specific small interfering RNA on increasing OC differentiation. In contrast to our findings regarding the RAW 264.7 cells, the ERK inhibitors attenuated the differentiation of bone marrow-derived cells into OCs. The ERK inhibitors significantly increased the phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK) but not the activation of p38 MAPK, Lyn, and mTOR. In addition, while the ERK inhibition increased the expression of the RANKL receptor RANK, it decreased the expression of negative mediators of OC differentiation, such as interferon regulatory factor-8, B-cell lymphoma 6, and interferon-γ. These dichotomous effects of ERK inhibition suggest that while ERKs may play positive roles in bone marrow-derived cells, ERKs may also play negative regulatory roles in RAW 264.7 cells. These data provide important information for drug development utilizing ERK inhibitors in OC-related disease treatment.
Collapse
Affiliation(s)
- Eun-Bi Choi
- Laboratory for Leukocyte Signaling Research, Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon 22212, Korea
- BK21 Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Korea
| | - Taiwo Samuel Agidigbi
- Laboratory for Leukocyte Signaling Research, Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon 22212, Korea
| | - In-Soon Kang
- Laboratory for Leukocyte Signaling Research, Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon 22212, Korea
- BK21 Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Korea
| | - Chaekyun Kim
- Laboratory for Leukocyte Signaling Research, Department of Pharmacology and Toxicology, College of Medicine, Inha University, Incheon 22212, Korea
- BK21 Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Korea
- Convergent Research Center for Metabolism and Immunoregulation, Inha University, Incheon 22212, Korea
| |
Collapse
|
7
|
Hong SJ, Jung S, Jang JS, Mo S, Kwon JO, Kim MK, Kim HH. PARK2 Induces Osteoclastogenesis through Activation of the NF-κB Pathway. Mol Cells 2022; 45:749-760. [PMID: 36047447 PMCID: PMC9589368 DOI: 10.14348/molcells.2022.0058] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 11/27/2022] Open
Abstract
Osteoclast generation from monocyte/macrophage lineage precursor cells needs to be tightly regulated to maintain bone homeostasis and is frequently over-activated in inflammatory conditions. PARK2, a protein associated with Parkinson's disease, plays an important role in mitophagy via its ubiquitin ligase function. In this study, we investigated whether PARK2 is involved in osteoclastogenesis. PARK2 expression was found to be increased during the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation. PARK2 gene silencing with siRNA significantly reduced osteoclastogenesis induced by RANKL, LPS (lipopolysaccharide), TNFα (tumor necrosis factor α), and IL-1β (interleukin-1β). On the other hand, overexpression of PARK2 promoted osteoclastogenesis. This regulation of osteoclastogenesis by PARK2 was mediated by IKK (inhibitory κB kinase) and NF-κB activation while MAPK (mitogen-activated protein kinases) activation was not involved. Additionally, administration of PARK2 siRNA significantly reduced osteoclastogenesis and bone loss in an in vivo model of inflammatory bone erosion. Taken together, this study establishes a novel role for PARK2 as a positive regulator in osteoclast differentiation and inflammatory bone destruction.
Collapse
Affiliation(s)
- Seo Jin Hong
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Suhan Jung
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Ji Sun Jang
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Shenzheng Mo
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Jun-Oh Kwon
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
8
|
Liang W, Chen Q, Cheng S, Wei R, Li Y, Yao C, Ouyang Z, Kang D, Chen A, Liu Z, Li K, Bai X, Li Q, Huang B. Skin chronological aging drives age-related bone loss via secretion of cystatin-A. NATURE AGING 2022; 2:906-922. [PMID: 37118283 DOI: 10.1038/s43587-022-00285-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 08/25/2022] [Indexed: 04/30/2023]
Abstract
Although clinical evidence has indicated an association between skin atrophy and bone loss during aging, their causal relationship and the underlying mechanisms are unknown. Here we show that premature skin aging drives bone loss in mice. We further identify that cystatin-A (Csta), a keratinocyte-enriched secreted factor, mediates the effect of skin on bone. Keratinocyte-derived Csta binds the receptor for activated C-kinase 1 in osteoblast and osteoclast progenitors, thus promoting their proliferation but inhibiting osteoclast differentiation. Csta secretion decreases with skin aging in both mice and humans, thereby causing senile osteoporosis by differentially decreasing the numbers of osteoblasts and osteoclasts. In contrast, topical application of calcipotriol stimulates Csta production in the epidermis and alleviates osteoporosis. These results reveal a mode of endocrine regulation of bone metabolism in the skin, and identify Csta as an epidermally derived hormone linking skin aging to age-related bone loss. Enhancers of skin Csta levels could serve as a potential topical drug for treatment of senile osteoporosis.
Collapse
Affiliation(s)
- Wenquan Liang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qingjing Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shasha Cheng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ruiming Wei
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yuejun Li
- Department of Orthopedics, The Second People's Hospital of Panyu District, Guangzhou, China
| | - Chenfeng Yao
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhicong Ouyang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dawei Kang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ajuan Chen
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zezheng Liu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Kai Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Qingchu Li
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Bin Huang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Matsubara T, Yasuda K, Mizuta K, Kawaue H, Kokabu S. Tyrosine Kinase Src Is a Regulatory Factor of Bone Homeostasis. Int J Mol Sci 2022; 23:ijms23105508. [PMID: 35628319 PMCID: PMC9146043 DOI: 10.3390/ijms23105508] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoclasts, which resorb the bone, and osteoblasts, which form the bone, are the key cells regulating bone homeostasis. Osteoporosis and other metabolic bone diseases occur when osteoclast-mediated bone resorption is increased and bone formation by osteoblasts is decreased. Analyses of tyrosine kinase Src-knockout mice revealed that Src is essential for bone resorption by osteoclasts and suppresses bone formation by osteoblasts. Src-knockout mice exhibit osteopetrosis. Therefore, Src is a potential target for osteoporosis therapy. However, Src is ubiquitously expressed in many tissues and is involved in various biological processes, such as cell proliferation, growth, and migration. Thus, it is challenging to develop effective osteoporosis therapies targeting Src. To solve this problem, it is necessary to understand the molecular mechanism of Src function in the bone. Src expression and catalytic activity are maintained at high levels in osteoclasts. The high activity of Src is essential for the attachment of osteoclasts to the bone matrix and to resorb the bone by regulating actin-related molecules. Src also inhibits the activity of Runx2, a master regulator of osteoblast differentiation, suppressing bone formation in osteoblasts. In this paper, we introduce the molecular mechanisms of Src in osteoclasts and osteoblasts to explore its potential for bone metabolic disease therapy.
Collapse
|
10
|
Lu D, Demissie S, Horowitz NB, Gower AC, Lenburg ME, Alekseyev YO, Hussein AI, Bragdon B, Liu Y, Daukss D, Page JM, Webster MZ, Schlezinger JJ, Morgan EF, Gerstenfeld LC. Temporal and Quantitative Transcriptomic Differences Define Sexual Dimorphism in Murine Postnatal Bone Aging. JBMR Plus 2021; 6:e10579. [PMID: 35229061 PMCID: PMC8861981 DOI: 10.1002/jbm4.10579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Time is a central element of the sexual dimorphic patterns of development, pathology, and aging of the skeleton. Because the transcriptome is a representation of the phenome, we hypothesized that both sex and sex‐specific temporal, transcriptomic differences in bone tissues over an 18‐month period would be informative to the underlying molecular processes that lead to postnatal sexual dimorphism. Regardless of age, sex‐associated changes of the whole bone transcriptomes were primarily associated not only with bone but also vascular and connective tissue ontologies. A pattern‐based approach used to screen the entire Gene Expression Omnibus (GEO) database against those that were sex‐specific in bone identified two coordinately regulated gene sets: one related to high phosphate–induced aortic calcification and one induced by mechanical stimulation in bone. Temporal clustering of the transcriptome identified two skeletal tissue‐associated, sex‐specific patterns of gene expression. One set of genes, associated with skeletal patterning and morphology, showed peak expression earlier in females. The second set of genes, associated with coupled remodeling, had quantitatively higher expression in females and exhibited a broad peak between 3 to 12 months, concurrent with the animals' reproductive period. Results of phenome‐level structural assessments of the tibia and vertebrae, and in vivo and in vitro analysis of cells having osteogenic potential, were consistent with the existence of functionally unique, skeletogenic cell populations that are separately responsible for appositional growth and intramedullary functions. These data suggest that skeletal sexual dimorphism arises through sex‐specific, temporally different processes controlling morphometric growth and later coupled remodeling of the skeleton during the reproductive period of the animal. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Darlene Lu
- Department of Biostatistics Boston University School of Public Health Boston MA USA
| | - Serkalem Demissie
- Department of Biostatistics Boston University School of Public Health Boston MA USA
| | - Nina B Horowitz
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| | - Adam C Gower
- Department of Medicine, Section of Computational Biomedicine Boston University School of Medicine Boston MA USA
| | - Marc E Lenburg
- Department of Medicine, Section of Computational Biomedicine Boston University School of Medicine Boston MA USA
- Department of Pathology and Laboratory Medicine Boston University School of Medicine Boston MA USA
| | - Yuriy O Alekseyev
- Department of Pathology and Laboratory Medicine Boston University School of Medicine Boston MA USA
| | - Amira I Hussein
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| | - Beth Bragdon
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| | - Yu Liu
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| | - Dana Daukss
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| | - Jack M Page
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| | - Micheal Z Webster
- Department of Mechanical Engineering Boston University Boston MA USA
| | - Jennifer J Schlezinger
- Department of Environmental Health Boston University School of Public Health Boston MA USA
| | - Elise F Morgan
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
- Department of Mechanical Engineering Boston University Boston MA USA
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery Boston University School of Medicine Boston MA USA
| |
Collapse
|
11
|
Bai B, Chen Q, Jing R, He X, Wang H, Ban Y, Ye Q, Xu W, Zheng C. Molecular Basis of Prostate Cancer and Natural Products as Potential Chemotherapeutic and Chemopreventive Agents. Front Pharmacol 2021; 12:738235. [PMID: 34630112 PMCID: PMC8495205 DOI: 10.3389/fphar.2021.738235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer is the second most common malignant cancer in males. It involves a complex process driven by diverse molecular pathways that closely related to the survival, apoptosis, metabolic and metastatic characteristics of aggressive cancer. Prostate cancer can be categorized into androgen dependent prostate cancer and castration-resistant prostate cancer and cure remains elusive due to the developed resistance of the disease. Natural compounds represent an extraordinary resource of structural scaffolds with high diversity that can offer promising chemical agents for making prostate cancer less devastating and curable. Herein, those natural compounds of different origins and structures with potential cytotoxicity and/or in vivo anti-tumor activities against prostate cancer are critically reviewed and summarized according to the cellular signaling pathways they interfere. Moreover, the anti-prostate cancer efficacy of many nutrients, medicinal plant extracts and Chinese medical formulations were presented, and the future prospects for the application of these compounds and extracts were discussed. Although the failure of conventional chemotherapy as well as involved serious side effects makes natural products ideal candidates for the treatment of prostate cancer, more investigations of preclinical and even clinical studies are necessary to make use of these medical substances reasonably. Therefore, the elucidation of structure-activity relationship and precise mechanism of action, identification of novel potential molecular targets, and optimization of drug combination are essential in natural medicine research and development.
Collapse
Affiliation(s)
- Bingke Bai
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Qianbo Chen
- Department of Anesthesiology, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Rui Jing
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xuhui He
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Hongrui Wang
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yanfei Ban
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Qi Ye
- Department of Biological Science, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Weiheng Xu
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Chengjian Zheng
- Department of Chinese Medicine Authentication, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
12
|
Kondo T, Otsuka Y, Aoki H, Goto Y, Kawaguchi Y, Waguri-Nagaya Y, Miyazawa K, Goto S, Aoyama M. The Inducible Nitric Oxide Synthase Pathway Promotes Osteoclastogenesis under Hypoxic Culture Conditions. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2072-2079. [PMID: 34560064 DOI: 10.1016/j.ajpath.2021.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/09/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022]
Abstract
Bone homeostasis depends on the balance between bone resorption by osteoclasts (OCs) and bone formation by osteoblasts. Bone resorption can become excessive under various pathologic conditions, including rheumatoid arthritis. Previous studies have shown that OC formation is promoted under hypoxia. However, the precise mechanisms behind OC formation under hypoxia have not been elucidated. The present study investigated the role of inducible nitric oxide synthase (iNOS) in OC differentiation under hypoxia. Primary bone marrow cells obtained from mice were stimulated with receptor activator of NF-κB ligand and macrophage colony-stimulating factor to induce OC differentiation. The number of OCs increased in culture under hypoxia (oxygen concentration, 5%) compared with that under normoxia (oxygen concentration, 20%). iNOS gene and protein expression increased in culture under hypoxia. Addition of an iNOS inhibitor under hypoxic conditions suppressed osteoclastogenesis. Addition of a nitric oxide donor to the normoxic culture promoted osteoclastogenesis. Furthermore, insulin-like growth factor 2 expression was significantly altered in both iNOS inhibition experiments and nitric oxide donor experiments. These data might provide clues to therapies for excessive osteoclastogenesis under several hypoxic pathologic conditions, including rheumatoid arthritis.
Collapse
Affiliation(s)
- Takao Kondo
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Yuto Otsuka
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Yoh Goto
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | - Yohei Kawaguchi
- Department of Glial Cell Biology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan; Department of Orthopaedic Surgery, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Yuko Waguri-Nagaya
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Ken Miyazawa
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | - Shigemi Goto
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan.
| |
Collapse
|
13
|
Yasuda K, Matsubara T, Shirakawa T, Kawamoto T, Kokabu S. Protein phosphatase 1 regulatory subunit 18 suppresses the transcriptional activity of NFATc1 via regulation of c-fos. Bone Rep 2021; 15:101114. [PMID: 34401407 PMCID: PMC8353383 DOI: 10.1016/j.bonr.2021.101114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/17/2021] [Accepted: 08/01/2021] [Indexed: 12/20/2022] Open
Abstract
The transcription factor NFATc1 and its binding partner AP-1 (a complex containing c-fos and c-Jun) play a central role in osteoclast differentiation. NFATc1 and AP-1 promote the expression of target genes such as Acp5, Ctsk and also auto-regulate NFATc1 expression as well. We previously reported that protein phosphatase 1 regulatory subunit 18 (PPP1r18) is a negative regulator of osteoclast bone resorption by inhibiting cell attachment to bone matrix. We also reported that PPP1r18 potentially regulates NFATc1 expression during osteoclast differentiation. To further explore this, in this study we have examined the effect of PPP1r18 on NFATc1 expression and activity by overexpressing PPP1r18 during the early stage of osteoclast differentiation. We found that PPP1r18 suppressed NFATc1 expression through inhibition of the transcriptional activity of NFATc1. Since PPP1r18 does not regulate NFATc1 directly, we next explored the involvement of AP-1. Our data showed that c-fos phosphorylation and nuclear localization were reduced by PPP1r18 overexpression. Further experiments showed that overexpression of c-fos together with PPP1r18 rescued NFATc1 expression and transcriptional activity. Moreover, c-fos activity inhibition by PPP1r18 was canceled by mutation of the phosphatase binding site of PPP1r18. Taken together, PPP1r18-regulated phosphatase activity targets c-fos phosphorylation and suppresses subsequent NFATc1 expression and activity. PPP1r18 suppresses osteoclast differentiation. PPP1r18 suppresses c-fos phosphorylation and nuclear localization. PPP1r18 suppresses NFAT via c-fos.
Collapse
Key Words
- Ctsk, cathepsin K
- Dc-stamp, dendrocyte expressed seven transmembrane protein
- GapDH, glyceraldehyde-3-phosphate dehydrogenase
- M-CSF, macrophage colony stimulating factor
- NFATc1
- NFATc1, nuclear factor of activated T cells 1
- Osteoclast
- PP1, protein phosphatase 1
- PPP1r18
- PPP1r18, protein phosphatase 1 regulatory subunit 18
- RANK, receptor activator nuclear factor kappa B
- RANKL, receptor activator nuclear factor kappa B ligand
- Src, Rous sarcoma oncogene
- TRAP, tartrate resistant acid phosphatase
- c-Fos
- c-Jun, Jun proto-oncogene, AP-1 transcription factor subunit
- c-fos, Fos proto-oncogene, AP-1 transcription factor subunit
Collapse
Affiliation(s)
- Kazuma Yasuda
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Corresponding authors.
| | - Tomohiko Shirakawa
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka 803-8580, Japan
- Corresponding authors.
| |
Collapse
|
14
|
Matsubara T, Addison WN, Kokabu S, Neff L, Horne W, Gori F, Baron R. Characterization of unique functionalities in c-Src domains required for osteoclast podosome belt formation. J Biol Chem 2021; 296:100790. [PMID: 34019873 PMCID: PMC8196221 DOI: 10.1016/j.jbc.2021.100790] [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] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/03/2021] [Accepted: 05/12/2021] [Indexed: 10/26/2022] Open
Abstract
Deletion of c-Src, a ubiquitously expressed tyrosine kinase, results in osteoclast dysfunction and osteopetrosis, in which bones harden into "stone." In contrast, deletion of the genes encoding other members of the Src family kinase (SFK) fails to produce an osteopetrotic phenotype. This suggests that c-Src performs a unique function in the osteoclast that cannot be compensated for by other SFKs. We aimed to identify the molecular basis of this unique role in osteoclasts and bone resorption. We found that c-Src, Lyn, and Fyn were the most highly expressed SFKs in WT osteoclasts, whereas Hck, Lck, Blk, and Fgr displayed low levels of expression. Formation of the podosome belt, clusters of unique actin assemblies, was disrupted in src-/- osteoclasts; introduction of constitutively activated SFKs revealed that only c-Src and Fyn could restore this process. To identify the key structural domains responsible, we constructed chimeric Src-Hck and Src-Lyn constructs in which the unique, SH3, SH2, or catalytic domains had been swapped. We found that the Src unique, SH3, and kinase domains were each crucial to establish Src functionality. The SH2 domain could however be substituted with Lyn or Hck SH2 domains. Furthermore, we demonstrate that c-Src's functionality is, in part, derived from an SH3-proximal proline-rich domain interaction with c-Cbl, leading to phosphorylation of c-Cbl Tyr700. These data help clarify Src's unique functionality in the organization of the cytoskeleton in osteoclasts, required for efficient bone resorption and explain why c-Src cannot be replaced, in osteoclasts, by other SFKs.
Collapse
Affiliation(s)
- Takuma Matsubara
- Division of Bone and Mineral Research, Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA; Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Fukuoka, Japan.
| | - William N Addison
- Division of Bone and Mineral Research, Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA; Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Fukuoka, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Fukuoka, Japan
| | - Lynn Neff
- Division of Bone and Mineral Research, Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - William Horne
- Division of Bone and Mineral Research, Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Francesca Gori
- Division of Bone and Mineral Research, Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Roland Baron
- Division of Bone and Mineral Research, Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School and Endocrine Unit, MGH, Boston, Massachusetts, USA.
| |
Collapse
|
15
|
Biological Effects of β-Glucans on Osteoclastogenesis. Molecules 2021; 26:molecules26071982. [PMID: 33915775 PMCID: PMC8036280 DOI: 10.3390/molecules26071982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/02/2022] Open
Abstract
Although the anti-tumor and anti-infective properties of β-glucans have been well-discussed, their role in bone metabolism has not been reviewed so far. This review discusses the biological effects of β-glucans on bone metabolisms, especially on bone-resorbing osteoclasts, which are differentiated from hematopoietic precursors. Multiple immunoreceptors that can recognize β-glucans were reported to be expressed in osteoclast precursors. Coordinated co-stimulatory signals mediated by these immunoreceptors are important for the regulation of osteoclastogenesis and bone remodeling. Curdlan from the bacterium Alcaligenes faecalis negatively regulates osteoclast differentiation in vitro by affecting both the osteoclast precursors and osteoclast-supporting cells. We also showed that laminarin, lichenan, and glucan from baker’s yeast, as well as β-1,3-glucan from Euglema gracilisas, inhibit the osteoclast formation in bone marrow cells. Consistent with these findings, systemic and local administration of β-glucan derived from Aureobasidium pullulans and Saccharomyces cerevisiae suppressed bone resorption in vivo. However, zymosan derived from S. cerevisiae stimulated the bone resorption activity and is widely used to induce arthritis in animal models. Additional research concerning the relationship between the molecular structure of β-glucan and its effect on osteoclastic bone resorption will be beneficial for the development of novel treatment strategies for bone-related diseases.
Collapse
|
16
|
Shalev M, Arman E, Stein M, Cohen-Sharir Y, Brumfeld V, Kapishnikov S, Royal I, Tuckermann J, Elson A. PTPRJ promotes osteoclast maturation and activity by inhibiting Cbl-mediated ubiquitination of NFATc1 in late osteoclastogenesis. FEBS J 2021; 288:4702-4723. [PMID: 33605542 DOI: 10.1111/febs.15778] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/22/2021] [Accepted: 02/18/2021] [Indexed: 11/28/2022]
Abstract
Bone-resorbing osteoclasts (OCLs) are multinucleated phagocytes, whose central roles in regulating bone formation and homeostasis are critical for normal health and development. OCLs are produced from precursor monocytes in a multistage process that includes initial differentiation, cell-cell fusion, and subsequent functional and morphological maturation; the molecular regulation of osteoclastogenesis is not fully understood. Here, we identify the receptor-type protein tyrosine phosphatase PTPRJ as an essential regulator specifically of OCL maturation. Monocytes from PTPRJ-deficient (JKO) mice differentiate and fuse normally, but their maturation into functional OCLs and their ability to degrade bone are severely inhibited. In agreement, mice lacking PTPRJ throughout their bodies or only in OCLs exhibit increased bone mass due to reduced OCL-mediated bone resorption. We further show that PTPRJ promotes OCL maturation by dephosphorylating the M-CSF receptor (M-CSFR) and Cbl, thus reducing the ubiquitination and degradation of the key osteoclastogenic transcription factor NFATc1. Loss of PTPRJ increases ubiquitination of NFATc1 and reduces its amounts at later stages of osteoclastogenesis, thereby inhibiting OCL maturation. PTPRJ thus fulfills an essential and cell-autonomous role in promoting OCL maturation by balancing between the pro- and anti-osteoclastogenic activities of the M-CSFR and maintaining NFATc1 expression during late osteoclastogenesis.
Collapse
Affiliation(s)
- Moran Shalev
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Esther Arman
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, University of Ulm, Germany
| | - Yael Cohen-Sharir
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, The Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Kapishnikov
- Department of Chemical Research Support, The Weizmann Institute of Science, Rehovot, Israel
| | - Isabelle Royal
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, QC, Canada.,Institut du Cancer de Montréal, QC, Canada.,Department of Medicine, University of Montreal, QC, Canada
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Germany
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
17
|
Imerb N, Thonusin C, Chattipakorn N, Chattipakorn SC. Aging, obese-insulin resistance, and bone remodeling. Mech Ageing Dev 2020; 191:111335. [DOI: 10.1016/j.mad.2020.111335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 08/14/2020] [Indexed: 02/08/2023]
|
18
|
Zhao Y, Jia L, Zheng Y, Li W. Involvement of Noncoding RNAs in the Differentiation of Osteoclasts. Stem Cells Int 2020; 2020:4813140. [PMID: 32908541 PMCID: PMC7468661 DOI: 10.1155/2020/4813140] [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: 02/21/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
As the most important bone-resorbing cells, osteoclasts play fundamental roles in bone remodeling and skeletal health. Much effort has been focused on identifying the regulators of osteoclast metabolism. Noncoding RNAs (ncRNAs) reportedly regulate osteoclast formation, differentiation, survival, and bone-resorbing activity to participate in bone physiology and pathology. The present review intends to provide a general framework for how ncRNAs and their targets regulate osteoclast differentiation and the important events of osteoclastogenesis they are involved in, including osteoclast precursor generation, early differentiation, mononuclear osteoclast fusion, and multinucleated osteoclast function and survival. This framework is beneficial for understanding bone biology and for identifying the potential biomarkers or therapeutic targets of bone diseases. The review also summarizes the results of in vivo experiments and classic experiment methods for osteoclast-related researches.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Lingfei Jia
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
19
|
Results of a Randomized, Double-Blinded, Placebo-Controlled, Phase 2.5 Study of Saracatinib (AZD0530), in Patients with Recurrent Osteosarcoma Localized to the Lung. Sarcoma 2020; 2020:7935475. [PMID: 32398945 PMCID: PMC7211262 DOI: 10.1155/2020/7935475] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose Osteosarcoma is a rare cancer and a third of patients who have completed primary treatment will develop osteosarcoma recurrence. The Src pathway has been implicated in the metastatic behavior of osteosarcoma; about 95% of samples examined express Src or have evidence of downstream activation of this pathway. Saracatinib (AZD0530) is a potent and selective Src kinase inhibitor that was evaluated in adults in Phase 1 studies. The primary goal of this study was to determine if treatment with saracatinib could increase progression-free survival (PFS) for patients who have undergone complete resection of osteosarcoma lung metastases in a double-blinded, placebo-controlled trial. Patients and Methods. Subjects with recurrent osteosarcoma localized to lung and who had complete surgical removal of all lung nodules were randomized within six weeks after complete surgical resection. Saracatinib, or placebo, was administered at a dose of 175 mg orally, once daily, for up to thirteen 28-day cycles. Results Thirty-seven subjects were included in the analyses; 18 subjects were randomized to receive saracatinib and 19 to receive placebo. Intent-to-treat analysis demonstrated a median PFS of 19.4 months in the saracatinib treatment group and 8.6 months in the placebo treatment group (p=0.47). Median OS was not reached in either arm. Conclusions Although saracatinib was well tolerated in this patient population, there was no apparent impact of the drug in this double-blinded, placebo-controlled trial on OS, and Src inhibition alone may not be sufficient to suppress metastatic progression in osteosarcoma. There is a suggestion of potential clinical benefit as evidenced by longer PFS in patients randomized to saracatinib based on limited numbers of patients treated.
Collapse
|
20
|
Matsubara T, Yaginuma T, Addison WN, Fujita Y, Watanabe K, Yoshioka I, Hikiji H, Maki K, Baron R, Kokabu S. Plectin stabilizes microtubules during osteoclastic bone resorption by acting as a scaffold for Src and Pyk2. Bone 2020; 132:115209. [PMID: 31866495 DOI: 10.1016/j.bone.2019.115209] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 10/25/2022]
Abstract
Osteoclasts are multinuclear cells which maintain bone homeostasis by resorbing bone. During bone resorption, osteoclasts attach to the bone matrix via a sealing zone formed by an actin ring. Rous sarcoma oncogene (Src) is essential for actin ring formation and bone resorption. Recently, we demonstrated that plectin, a cytolinker protein, is a Src-binding protein in osteoclasts. However, the function of plectin in osteoclasts remains unknown. In this study, we demonstrated that shRNA knockdown of plectin in RAW 264.7 cells resulted in tartrate resistant acid phosphatase positive multinuclear cells (TRAP (+) MNCs) with impaired actin ring formation and bone resorption activity. Moreover, we found that in plectin-silenced TRAP (+) MNCs, Src and protein tyrosine kinase 2 beta (Pyk2), two critical kinases in osteoclastic bone resorption, were inactivated and microtubule polarity was disturbed. These results suggest that plectin plays a critical role in osteoclast biology by acting as a scaffold to facilitate Src and Pyk2 activation during microtubule organization.
Collapse
Affiliation(s)
- Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan.
| | - Tatsuki Yaginuma
- Division of Molecular Signaling and Biochemistry, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan; Division of Oral Medicine, Department of Physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | - William N Addison
- Division of Molecular Signaling and Biochemistry, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan
| | - Yuko Fujita
- Division of Developmental Stomatognathic Function Science, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan
| | - Kouji Watanabe
- Division of Developmental Stomatognathic Function Science, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan
| | - Izumi Yoshioka
- Division of Oral Medicine, Department of Physical Functions, Kyushu Dental University, Kitakyushu, Japan
| | - Hisako Hikiji
- School of Oral Health Sciences, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan
| | - Kenshi Maki
- Division of Developmental Stomatognathic Function Science, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan
| | - Roland Baron
- Department of Medicine, Harvard Medical School and Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, 188 Longwood Ave., Boston, MA 02115, USA
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Promotion, Kyushu Dental University, 2-6-1, Manazuru, Kitakyushu, Fukuoka 8038580, Japan
| |
Collapse
|
21
|
Raynaud-Messina B, Verollet C, Maridonneau-Parini I. The osteoclast, a target cell for microorganisms. Bone 2019; 127:315-323. [PMID: 31233933 DOI: 10.1016/j.bone.2019.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/11/2019] [Accepted: 06/21/2019] [Indexed: 02/02/2023]
Abstract
Bone is a highly adaptive tissue with regenerative properties that is subject to numerous diseases. Infection is one of the causes of altered bone homeostasis. Bone infection happens subsequently to bone surgery or to systemic spreading of microorganisms. In addition to osteoblasts, osteoclasts (OCs) also constitute cell targets for pathogens. OCs are multinucleated cells that have the exclusive ability to resorb bone mineral tissue. However, the OC is much more than a bone eater. Beyond its role in the control of bone turnover, the OC is an immune cell that produces and senses inflammatory cytokines, ingests microorganisms and presents antigens. Today, increasing evidence shows that several pathogens use OC as a host cell to grow, generating debilitating bone defects. In this review, we exhaustively inventory the bacteria and viruses that infect OC and report the present knowledge in this topic. We point out that most of the microorganisms enhance the bone resorption activity of OC. We notice that pathogen interactions with the OC require further investigation, in particular to validate the OC as a host cell in vivo and to identify the cellular mechanisms involved in altered bone resorption. Thus, we conclude that the OC is a new cell target for pathogens; this new research area paves the way for new therapeutic strategies in the infections causing bone defects.
Collapse
Affiliation(s)
- Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France; International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| |
Collapse
|
22
|
Erami Z, Heitz S, Bresnick AR, Backer JM. PI3Kβ links integrin activation and PI(3,4)P 2 production during invadopodial maturation. Mol Biol Cell 2019; 30:2367-2376. [PMID: 31318314 PMCID: PMC6741064 DOI: 10.1091/mbc.e19-03-0182] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/17/2019] [Accepted: 07/01/2019] [Indexed: 11/17/2022] Open
Abstract
The invasion of tumor cells from the primary tumor is mediated by invadopodia, actin-rich protrusive organelles that secrete matrix metalloproteases and degrade the extracellular matrix. This coupling between protrusive activity and matrix degradation facilitates tumor invasion. We previously reported that the PI3Kβ isoform of PI 3-kinase, which is regulated by both receptor tyrosine kinases and G protein-coupled receptors, is required for invasion and gelatin degradation in breast cancer cells. We have now defined the mechanism by which PI3Kβ regulates invadopodia. We find that PI3Kβ is specifically activated downstream from integrins, and is required for integrin-stimulated spreading and haptotaxis as well as integrin-stimulated invadopodia formation. Surprisingly, these integrin-stimulated and PI3Kβ-dependent responses require the production of PI(3,4)P2 by the phosphoinositide 5'-phosphatase SHIP2. Thus, integrin activation of PI3Kβ is coupled to the SHIP2-dependent production of PI(3,4)P2, which regulates the recruitment of PH domain-containing scaffolds such as lamellipodin to invadopodia. These findings provide novel mechanistic insight into the role of PI3Kβ in the regulation of invadopodia in breast cancer cells.
Collapse
Affiliation(s)
- Zahra Erami
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Samantha Heitz
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Anne R. Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jonathan M. Backer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
23
|
Dewulf J, Vangestel C, Verhoeven Y, van Dam P, Elvas F, Van den Wyngaert T, Clézardin P. Bone metastases in the era of targeted treatments: insights from molecular biology. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:98-111. [PMID: 31298015 DOI: 10.23736/s1824-4785.19.03203-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bone metastases remain a common feature of advanced cancers and are associated with significant morbidity and mortality. Recent research has identified promising novel treatment targets to improve current treatment strategies for bone metastatic disease. This review summarizes the well-known and recently discovered molecular biology pathways in bone that govern normal physiological remodeling or drive the pathophysiological changes observed when bone metastases are present. In the rapidly changing world of targeted cancer treatments, it is important to recognize the specific treatment effects induced in bone by these agents and the potential impact on common imaging strategies. The osteoclastic targets (bisphosphonates, LGR4, RANKL, mTOR, MET-VEGFR, cathepsin K, Src, Dock 5) and the osteoblastic targets (Wnt and endothelin) are discussed, and the emerging field of osteo-immunity is introduced as potential future therapeutic target. Finally, a summary is provided of available trial data for agents that target these pathways and that have been assessed in patients. The ultimate goal of research into novel pathways and targets involved in the tumor-bone microenvironment is to tackle one of the great remaining unmet needs in oncology, that is finding a cure for bone metastatic disease.
Collapse
Affiliation(s)
- Jonatan Dewulf
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Yannick Verhoeven
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Peter van Dam
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium.,Multidisciplinary Oncologic Center Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Filipe Elvas
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Tim Van den Wyngaert
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium - .,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Philippe Clézardin
- INSERM Laboratory Pathophysiology, Diagnosis and Treatments of Bone Diseases, Lyon, France.,INSERM European Associated Laboratory Cancer and Bone Metastasis, University of Sheffield, Medical School, Sheffield, UK
| |
Collapse
|
24
|
Narahara S, Sakai E, Kadowaki T, Yamaguchi Y, Narahara H, Okamoto K, Asahina I, Tsukuba T. KBTBD11, a novel BTB-Kelch protein, is a negative regulator of osteoclastogenesis through controlling Cullin3-mediated ubiquitination of NFATc1. Sci Rep 2019; 9:3523. [PMID: 30837587 PMCID: PMC6401029 DOI: 10.1038/s41598-019-40240-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/12/2019] [Indexed: 01/10/2023] Open
Abstract
Kelch repeat and BTB domain-containing protein 11 (KBTBD11) is a member of the KBTBD subfamily of proteins that possess a BTB domain and Kelch repeats. Despite the presence of the Kbtbd11 gene in mammalian genomes, there are few reports about KBTBD11 at present. In this study, we identified the novel protein KBTBD11 as a negative regulator of osteoclast differentiation. We found that expression of KBTBD11 increased during osteoclastogenesis. Small-interfering-RNA-mediated knockdown of KBTBD11 enhanced osteoclast formation, and markedly increased the expression of several osteoclast marker genes compared with control cells. Conversely, KBTBD11 overexpression impaired osteoclast differentiation, and decreased the expression of osteoclast marker genes. Among six major signaling pathways regulating osteoclast differentiation, KBTBD11 predominantly influenced the nuclear factor of activated T cell cytoplasmic-1 (NFATc1) pathway. Mechanistically, KBTBD11 was found to interact with an E3 ubiquitin ligase, Cullin3. Further experiments involving immunoprecipitation and treatment with MG132, a proteasome inhibitor, showed that the KBTBD11–Cullin3 promotes ubiquitination and degradation of NFATc1 by the proteasome. Considering that NFATc1 is an essential factor for osteoclast differentiation, the KBTBD11 and Cullin3 probably regulate the levels of NFATc1 through the ubiquitin-proteasome degradation system. Thus, KBTBD11 negatively modulates osteoclast differentiation by controlling Cullin3-mediated ubiquitination of NFATc1.
Collapse
Affiliation(s)
- Shun Narahara
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan.,Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Tomoko Kadowaki
- Department of Frontier Life Science, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Yu Yamaguchi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Haruna Narahara
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan.,Department of Dental Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8525, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan.
| |
Collapse
|
25
|
Plotkin LI, Bruzzaniti A. Molecular signaling in bone cells: Regulation of cell differentiation and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 116:237-281. [PMID: 31036293 PMCID: PMC7416488 DOI: 10.1016/bs.apcsb.2019.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The achievement of proper bone mass and architecture, and their maintenance throughout life requires the concerted actions of osteoblasts, the bone forming cells, and osteoclasts, the bone resorbing cells. The differentiation and activity of osteoblasts and osteoclasts are regulated by molecules produced by matrix-embedded osteocytes, as well as by cross talk between osteoblasts and osteoclasts through secreted factors. In addition, it is likely that direct contact between osteoblast and osteoclast precursors, and the contact of these cells with osteocytes and cells in the bone marrow, also modulates bone cell differentiation and function. With the advancement of molecular and genetic tools, our comprehension of the intracellular signals activated in bone cells has evolved significantly, from early suggestions that osteoblasts and osteoclasts have common precursors and that osteocytes are inert cells in the bone matrix, to the very sophisticated understanding of a network of receptors, ligands, intracellular kinases/phosphatases, transcription factors, and cell-specific genes that are known today. These advances have allowed the design and FDA-approval of new therapies to preserve and increase bone mass and strength in a wide variety of pathological conditions, improving bone health from early childhood to the elderly. We have summarized here the current knowledge on selected intracellular signal pathways activated in osteoblasts, osteocytes, and osteoclasts.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| | - Angela Bruzzaniti
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States; Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, United States
| |
Collapse
|
26
|
Roth L, Wakim J, Wasserman E, Shalev M, Arman E, Stein M, Brumfeld V, Sagum CA, Bedford MT, Tuckermann J, Elson A. Phosphorylation of the phosphatase PTPROt at Tyr 399 is a molecular switch that controls osteoclast activity and bone mass in vivo. Sci Signal 2019; 12:12/563/eaau0240. [PMID: 30622194 DOI: 10.1126/scisignal.aau0240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Bone resorption by osteoclasts is essential for bone homeostasis. The kinase Src promotes osteoclast activity and is activated in osteoclasts by the receptor-type tyrosine phosphatase PTPROt. In other contexts, however, PTPROt can inhibit Src activity. Through in vivo and in vitro experiments, we show that PTPROt is bifunctional and can dephosphorylate Src both at its inhibitory residue Tyr527 and its activating residue Tyr416 Whereas wild-type and PTPROt knockout mice exhibited similar bone masses, mice in which a putative C-terminal phosphorylation site, Tyr399, in endogenous PTPROt was replaced with phenylalanine had increased bone mass and reduced osteoclast activity. Osteoclasts from the knock-in mice also showed reduced Src activity. Experiments in cultured cells and in osteoclasts derived from both mouse strains demonstrated that the absence of phosphorylation at Tyr399 caused PTPROt to dephosphorylate Src at the activating site pTyr416 In contrast, phosphorylation of PTPROt at Tyr399 enabled PTPROt to recruit Src through Grb2 and to dephosphorylate Src at the inhibitory site Tyr527, thus stimulating Src activity. We conclude that reversible phosphorylation of PTPROt at Tyr399 is a molecular switch that selects between its opposing activities toward Src and maintains a coherent signaling output, and that blocking this phosphorylation event can induce physiological effects in vivo. Because most receptor-type tyrosine phosphatases contain potential phosphorylation sites at their C termini, we propose that preventing phosphorylation at these sites or its consequences may offer an alternative to inhibiting their catalytic activity to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Lee Roth
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Jean Wakim
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elad Wasserman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Moran Shalev
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Esther Arman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm 89081, Germany
| | - Vlad Brumfeld
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Cari A Sagum
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm 89081, Germany
| | - Ari Elson
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
27
|
Abstract
Bone tissue is comprised of a collagen-rich matrix containing non-collagenous organic compounds, strengthened by mineral crystals. Bone strength reflects the amount and structure of bone, as well as its quality. These qualities are determined and maintained by osteoblasts (bone-forming cells) and osteoclasts (bone-resorbing cells) on the surface of the bone and osteocytes embedded within the bone matrix. Bone development and growth also involves cartilage cells (chondrocytes). These cells do not act in isolation, but function in a coordinated manner, including co-ordination within each lineage, between the cells of bone, and between these cells and other cell types within the bone microenvironment. This chapter will briefly outline the cells of bone, their major functions, and some communication pathways responsible for controlling bone development and remodeling.
Collapse
Affiliation(s)
- Niloufar Ansari
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Natalie A Sims
- Bone Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
28
|
B7-H3 promotes multiple myeloma cell survival and proliferation by ROS-dependent activation of Src/STAT3 and c-Cbl-mediated degradation of SOCS3. Leukemia 2018; 33:1475-1486. [PMID: 30573782 DOI: 10.1038/s41375-018-0331-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 10/17/2018] [Accepted: 11/16/2018] [Indexed: 12/16/2022]
Abstract
B7-H3 (CD276) is broadly overexpressed by multiple human cancers. It plays a vital role in tumor progression and has been accepted as one of the inhibitory B7 family checkpoint molecules. To identify the functions and underlying mechanisms of B7-H3 in multiple myeloma, we analyzed B7-H3 expression in myeloma patients and used siRNAs and overexpression plasmid of B7-H3 to investigate its roles and downstream signaling molecules in myeloma cell lines. The results showed that surface expression of B7-H3 was upregulated in myeloma samples and cell lines. Lower expression of B7-H3 in myeloma cells was associated with better progression-free survival. Myeloma cell survival, drug resistance, and tumor growth could be promoted by B7-H3. The molecular basis for these functional roles of B7-H3 involved the activation of JAK2/STAT3 via redox-mediated oxidation and activation of Src. We further identified a STAT3-promoting signaling pathway by which oxidant-mediated Src phosphorylation led to secondary activation of the E3 ubiquitin ligase c-Cbl. Activated c-Cbl subsequently caused specific proteasomal degradation of SOCS3, a negative regulator of JAK2/STAT3. These data indicate B7-H3's important role in the activation of ROS/Src/c-Cbl pathway in multiple myeloma which integrates redox regulation and sustained STAT3 activation at the level of degradation of STAT3 suppressor.
Collapse
|
29
|
Kim JM, Lee K, Jeong D. Selective regulation of osteoclast adhesion and spreading by PLCγ/PKCα-PKCδ/RhoA-Rac1 signaling. BMB Rep 2018; 51:230-235. [PMID: 29301608 PMCID: PMC5988577 DOI: 10.5483/bmbrep.2018.51.5.198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Indexed: 01/16/2023] Open
Abstract
Bone resorption by multinucleated osteoclasts is a multistep process involving adhesion to the bone matrix, migration to resorption sites, and formation of sealing zones and ruffled borders. Macrophage colony-stimulating factor (M-CSF) and osteopontin (OPN) have been shown to be involved in the bone resorption process by respective activation of integrin αvβ3 via “inside-out” and “outside-in” signaling. In this study, we investigated the link between signal modulators known to M-CSF- and OPN-induced osteoclast adhesion and spreading. M-CSF- and OPN-induced osteoclast adhesion was achieved via activation of stepwise signals, including integrin αvβ3, PLCγ, PKCδ, and Rac1. Osteoclast spreading induced by M-CSF and OPN was shown to be controlled via sequential activation, consistent with the osteoclast adhesion processes. In contrast to osteoclast adhesion, osteoclast spreading induced by M-CSF and OPN was blocked via activation of PLCγ/PKCα/RhoA signaling. The combined results indicate that osteoclast adhesion and spreading are selectively regulated via PLCγ/PKCα-PKCδ/RhoA-Rac1 signaling.
Collapse
Affiliation(s)
- Jin-Man Kim
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415; Asan Medical Center, Asan Institute for Life Sciences, Seoul 26493, Korea
| | - Kyunghee Lee
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Daewon Jeong
- Department of Microbiology, Laboratory of Bone Metabolism and Control, Yeungnam University College of Medicine, Daegu 42415, Korea
| |
Collapse
|
30
|
Ku B, Yun HY, Lee KW, Shin HC, Lee SR, Kim CH, Park H, Yi KY, Lee CH, Kim SJ. Identification of N-(5-(phenoxymethyl)-1,3,4-thiadiazol-2-yl)acetamide derivatives as novel protein tyrosine phosphatase epsilon inhibitors exhibiting anti-osteoclastic activity. Bioorg Med Chem 2018; 26:5204-5211. [PMID: 30249496 DOI: 10.1016/j.bmc.2018.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 01/06/2023]
Abstract
Cytosolic protein tyrosine phosphatase epsilon (cyt-PTPε) plays a central role in controlling differentiation and function of osteoclasts, whose overactivation causes osteoporosis. Based on our previous study reporting a number of cyt-PTPε inhibitory chemical compounds, we carried out a further and extended analysis of our compounds to examine their effects on cyt-PTPε-mediated dephosphorylation and on osteoclast organization and differentiation. Among five compounds showing target selectivity to cyt-PTPε over three other phosphatases in vitro, two compounds exhibited an inhibitory effect against the dephosphorylation of cellular Src protein, the cyt-PTPε substrate. Moreover, these two compounds caused destabilization of the podosome structure that is necessary for the bone-resorbing activity of osteoclasts, and also attenuated cellular differentiation of monocytes into osteoclasts, without affecting cell viability. Therefore, these findings not only verified anti-osteoclastic effects of our cyt-PTPε inhibitory compounds, but also showed that cyt-PTPε expressed in osteoclasts could be a putative therapeutic target worth considering.
Collapse
Affiliation(s)
- Bonsu Ku
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Bioscience, University of Science and Technology KRIBB School, Daejeon 34113, Republic of Korea
| | - Hye-Yeoung Yun
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Bioscience, University of Science and Technology KRIBB School, Daejeon 34113, Republic of Korea
| | - Kyung Won Lee
- Center for Information-Based Drug Research, Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Ho-Chul Shin
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Chang Hyen Kim
- Department of Oral and Maxillofacial Surgery, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hwangseo Park
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Kyu Yang Yi
- Center for Information-Based Drug Research, Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Chang Hoon Lee
- Center for Information-Based Drug Research, Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Seung Jun Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Bioscience, University of Science and Technology KRIBB School, Daejeon 34113, Republic of Korea.
| |
Collapse
|
31
|
Kobayashi Y, Uehara S, Udagawa N. Roles of non-canonical Wnt signaling pathways in bone resorption. J Oral Biosci 2018. [DOI: 10.1016/j.job.2018.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
32
|
A novel miR17/protein tyrosine phosphatase-oc/EphA4 regulatory axis of osteoclast activity. Arch Biochem Biophys 2018; 650:30-38. [PMID: 29763590 PMCID: PMC5985224 DOI: 10.1016/j.abb.2018.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 12/17/2022]
Abstract
Information about the molecular mechanisms leading to the activation of the osteoclast is relatively limited. While there is compelling evidence that the signaling mechanisms of Src and integrin β3 are essential for osteoclast activation, the regulation of these two signaling mechanisms is not fully understood. In this review, evidence supporting a novel regulatory axis of osteoclast activation that plays an upstream regulatory role in both the Src and integrin β3 signaling during osteoclast activation is discussed. This regulatory axis contains three unique components: a structurally unique transmembrane protein-tyrosine phosphatase, PTP-oc, EphA4, and miR17. In the first component, PTP-oc activates the Src signaling through dephosphorylation of the inhibitory tyr-527 of Src. This in turn activates the integrin β3 signaling, enhances the JNK2/NFκB signaling, promotes the ITAM/Syk signaling, and suppresses the ITIM/Shp1 signaling; the consequence of which is activation of the osteoclast. In the second component, EphA4 inhibits osteoclast activity by suppressing the integrin β3 signaling. PTP-oc relieves the suppressive actions of EphA4 by directly dephosphorylating EphA4. In the third component, PTP-oc expression is negatively regulated by miR17. Accordingly, suppression of miR17 during osteoclast activation upregulates the PTP-oc signaling and suppresses the EphA4 signaling, resulting in the activation of the osteoclast. This regulatory axis is unique, in that each of the three components acts to exert suppressive action on their respective immediate downstream inhibitory step. Because the final downstream event is the EphA4-mediated inhibition of osteoclast activation, the overall effect of this mechanism is the stimulation of osteoclast activity.
Collapse
|
33
|
Sousa S, Clézardin P. Bone-Targeted Therapies in Cancer-Induced Bone Disease. Calcif Tissue Int 2018; 102:227-250. [PMID: 29079995 DOI: 10.1007/s00223-017-0353-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/19/2017] [Indexed: 01/14/2023]
Abstract
Cancer-induced bone disease is a major source of morbidity and mortality in cancer patients. Thus, effective bone-targeted therapies are essential to improve disease-free, overall survival and quality of life of cancer patients with bone metastases. Depending of the cancer-type, bone metastases mainly involve the modulation of osteoclast and/or osteoblast activity by tumour cells. To inhibit metastatic bone disease effectively, it is imperative to understand its underlying mechanisms and identify the target cells for therapy. If the aim is to prevent bone metastasis, it is essential to target not only bone metastatic features in the tumour cells, but also tumour-nurturing bone microenvironment properties. The currently available bone-targeted agents mainly affect osteoclasts, inhibiting bone resorption (e.g. bisphosphonates, denosumab). Some agents targeting osteoblasts begin to emerge which target osteoblasts (e.g. romosozumab), activating bone formation. Moreover, certain drugs initially thought to target only osteoclasts are now known to have a dual action (activating osteoblasts and inhibiting osteoclasts, e.g. proteasome inhibitors). This review will focus on the evolution of bone-targeted therapies for the treatment of cancer-induced bone disease, summarizing preclinical and clinical findings obtained with anti-resorptive and bone anabolic therapies.
Collapse
Affiliation(s)
- Sofia Sousa
- National Institute of Health and Medical Research (INSERM), UMR 1033, 69372, Lyon, France.
- Faculty of Medicine Laennec, University of Lyon-1, 69372, Villeurbanne, France.
| | - Philippe Clézardin
- National Institute of Health and Medical Research (INSERM), UMR 1033, 69372, Lyon, France
- Faculty of Medicine Laennec, University of Lyon-1, 69372, Villeurbanne, France
- European Cancer and Bone Metastasis Laboratory, Department of Bone Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
34
|
Nihal M, Wood GS. c-CBL regulates melanoma proliferation, migration, invasion and the FAK-SRC-GRB2 nexus. Oncotarget 2018; 7:53869-53880. [PMID: 27472394 PMCID: PMC5288227 DOI: 10.18632/oncotarget.10861] [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: 04/05/2016] [Accepted: 07/10/2016] [Indexed: 12/28/2022] Open
Abstract
Melanoma is one of the most aggressive and lethal forms of skin cancer. Despite recent improvements in targeted therapies, many patients with advanced disease fail to achieve lasting tumor regression. Therefore, it is important to develop novel druggable targets that can be exploited to improve clinical outcome. Here, we studied the role of Casitas B-lineage lymphoma (c-CBL), an E3 ubiquitin ligase, in human melanoma. Employing quantitative real-time PCR and Western blot analysis in a panel of human melanoma cell lines (A375, G361, Hs-294T, SK-Mel-2, SK-Mel-28 and 451Lu), we found that c-CBL is strongly expressed in human melanoma cells at the mRNA and protein levels. Further, we determined c-CBL levels in clinical samples of melanomas and benign melanocytic nevi, using quantitative Nuance multispectral imaging. Compared to benign nevi, melanomas showed an overlapping range of c-CBL immunoreactivity. Small interfering RNA (siRNA)-mediated knockdown of c-CBL resulted in decreased proliferation, clonogenic survival and migration of melanoma cells. Furthermore, it also resulted in decreased cellular invasion in a 3D spheroid assay system. C-CBL and FAK are regulated by SRC, and FAK binds SRC and GRB2. C-CBL E3 ligase domain regulates receptor tyrosine kinase internalization through ubiquitination and its ring finger domain stabilizes the FAK-SRC-actin cytoskeleton thereby promoting cellular motility. C-CBL knockdown was associated with decreased protein and/or mRNA levels of SRC, FAK and GRB2. Taken together, we have provided evidence that c-CBL plays a role in melanoma cell proliferation, migration and invasion as well as inhibition of the FAK-GRB2-SRC nexus. Our findings indicate that additional studies are warranted to further dissect the role of c-CBL in melanoma and determine the therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Minakshi Nihal
- Department of Dermatology, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Paul P. Carbone Comprehensive Cancer Center, Madison, Wisconsin, USA
| | - Gary S Wood
- Department of Dermatology, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Paul P. Carbone Comprehensive Cancer Center, Madison, Wisconsin, USA.,Wm. S. Middleton VA Medical Centre, Madison, Wisconsin, USA
| |
Collapse
|
35
|
The Actin-Binding Protein PPP1r18 Regulates Maturation, Actin Organization, and Bone Resorption Activity of Osteoclasts. Mol Cell Biol 2018; 38:MCB.00425-17. [PMID: 29158294 DOI: 10.1128/mcb.00425-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/11/2017] [Indexed: 01/21/2023] Open
Abstract
Osteoclasts resorb bone by attaching on the bone matrix and forming a sealing zone. In Src-deficient mice, osteoclasts cannot form the actin ring, a characteristic actin structure that seals the resorbed area, and resorb hardly any bone as a result. However, the molecular mechanism underlying the role of Src in the regulation and organization of the actin ring is still unclear. We identified an actin-regulatory protein, protein phosphatase 1 regulatory subunit 18 (PPP1r18), as an Src-binding protein in an Src-, Yes-, and Fyn-deficient fibroblast (SYF) cell line overexpressing a constitutively active form of Src. PPP1r18 was localized in the nucleus and actin ring. PPP1r18 overexpression in osteoclasts inhibited terminal differentiation, actin ring formation, and bone-resorbing activity. A mutation of the protein phosphatase 1 (PP1)-binding domain of PPP1r18 rescued these phenotypes. In contrast, PPP1r18 knockdown promoted terminal differentiation and actin ring formation. In summary, we showed that PPP1r18 likely plays a role in podosome organization and bone resorption.
Collapse
|
36
|
Heusschen R, Muller J, Binsfeld M, Marty C, Plougonven E, Dubois S, Mahli N, Moermans K, Carmeliet G, Léonard A, Baron F, Beguin Y, Menu E, Cohen-Solal M, Caers J. SRC kinase inhibition with saracatinib limits the development of osteolytic bone disease in multiple myeloma. Oncotarget 2017; 7:30712-29. [PMID: 27095574 PMCID: PMC5058712 DOI: 10.18632/oncotarget.8750] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/31/2016] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma (MM)-associated osteolytic bone disease is a major cause of morbidity and mortality in MM patients and the development of new therapeutic strategies is of great interest. The proto-oncogene SRC is an attractive target for such a strategy. In the current study, we investigated the effect of treatment with the SRC inhibitor saracatinib (AZD0530) on osteoclast and osteoblast differentiation and function, and on the development of MM and its associated bone disease in the 5TGM.1 and 5T2MM murine MM models. In vitro data showed an inhibitory effect of saracatinib on osteoclast differentiation, polarization and resorptive function. In osteoblasts, collagen deposition and matrix mineralization were affected by saracatinib. MM cell proliferation and tumor burden remained unaltered following saracatinib treatment and we could not detect any synergistic effects with drugs that are part of standard care in MM. We observed a marked reduction of bone loss after treatment of MM-bearing mice with saracatinib as reflected by a restoration of trabecular bone parameters to levels observed in naive control mice. Histomorphometric analyses support that this occurs through an inhibition of bone resorption. In conclusion, these data further establish SRC inhibition as a promising therapeutic approach for the treatment of MM-associated osteolytic bone disease.
Collapse
Affiliation(s)
- Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Joséphine Muller
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Marilène Binsfeld
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Caroline Marty
- INSERM-UMR-1132, Hôpital Lariboisière and Université Paris Diderot, Paris, France
| | - Erwan Plougonven
- Department of Chemical Engineering, PEPs (Products, Environments, Processes), University of Liège, Liège, Belgium
| | - Sophie Dubois
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Nadia Mahli
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Karen Moermans
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Angélique Léonard
- Department of Chemical Engineering, PEPs (Products, Environments, Processes), University of Liège, Liège, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium.,Division of Hematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium.,Division of Hematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Martine Cohen-Solal
- INSERM-UMR-1132, Hôpital Lariboisière and Université Paris Diderot, Paris, France
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium.,Division of Hematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| |
Collapse
|
37
|
Abstract
C–C chemokine receptor 5 (CCR5) is a co-receptor of HIV. Epidemiological findings suggest that the functional loss of CCR5 is correlated with a lower incidence of bone-destructive diseases as well as of HIV transmission. However, it is not clear whether CCR5 is involved in regulation of the function of bone cells, in addition to that of immune cells. Here we show that blockade of CCR5 using specific antibodies impairs human osteoclast function in vitro. Ccr5-deficient (Ccr5−/−) mice presented with dysfunctional osteoclasts and were resistant to osteoporosis induced by receptor activator of nuclear factor kappa-B ligand (RANKL), which triggers osteoporosis independently of inflammatory and immunomodulatory pathways. Furthermore, Ccr5 deficiency impairs the cellular locomotion and bone-resorption activity of osteoclasts, which is associated with the disarrangement of podosomes and adhesion complex molecules including Pyk2. Overall, the data provides evidence that CCR5 has an essential role in bone-destructive conditions through the functional regulation of osteoclasts. CCR5 is a co-receptor for HIV, and loss of function is associated with lower incidence of HIV but also with bone-destructive diseases. Here the authors show that ablation of CCR5 impairs osteoclast function and improves resistance to osteoporosis in mouse models.
Collapse
|
38
|
Liu X, Chin JF, Qu X, Bi H, Liu Y, Yu Z, Zhai Z, Qin A, Zhang B, Dai M. The Beneficial Effect of Praeruptorin C on Osteoporotic Bone in Ovariectomized Mice via Suppression of Osteoclast Formation and Bone Resorption. Front Pharmacol 2017; 8:627. [PMID: 28955232 PMCID: PMC5601062 DOI: 10.3389/fphar.2017.00627] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/28/2017] [Indexed: 01/23/2023] Open
Abstract
Being a highly prevalent disease, osteoporosis causes metabolism defects. Low bone density, compromised bone strength, and an increased danger of fragility fracture are its main characteristics. Natural compounds have been considered as potential alternative therapeutic agents for treating osteoporosis. In this study, we demonstrated that a natural compound, praeruptorin C (Pra-C), derived from the dried roots of Peucedanum praeruptorum, has beneficial effects in suppressing osteoclast formation and resorption function via attenuating the activation of nuclear factor kappa B as well as c-Jun N-terminal kinase/mitogen-activated protein kinase signaling pathways. Moreover, Pra-C was tested in the ovariectomized (OVX) mice, a well-established model of post-menopausal bone loss, and the results indicated Pra-C exerted beneficial effects on inhibiting excessive osteoclast activity and increasing bone mass of OVX mice. Therefore, the protective effects of Pra-C on OVX mice bone are related to its inhibition of osteoclast formation and bone resorption, suggesting that Pra-C is a good potential candidate for osteoporosis treatment.
Collapse
Affiliation(s)
- Xuqiang Liu
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang, China
| | - Jie-Fen Chin
- Department of Orthopedics, Erasmus University Medical CenterRotterdam, Netherlands
| | - Xinhua Qu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Haidi Bi
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang, China
| | - Yuan Liu
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang, China
| | - Ziqiang Yu
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang, China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Bin Zhang
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang, China
| | - Min Dai
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang, China
| |
Collapse
|
39
|
Uehara S, Udagawa N, Mukai H, Ishihara A, Maeda K, Yamashita T, Murakami K, Nishita M, Nakamura T, Kato S, Minami Y, Takahashi N, Kobayashi Y. Protein kinase N3 promotes bone resorption by osteoclasts in response to Wnt5a-Ror2 signaling. Sci Signal 2017; 10:10/494/eaan0023. [DOI: 10.1126/scisignal.aan0023] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Liu X, Qu X, Nie T, Zhai Z, Li H, Ouyang Z, Qin A, Zhang S, Zhang S, Fan Q, Tang T, Yu Z, Dai M. The Beneficial Effects of Bisphosphonate-enoxacin on Cortical Bone Mass and Strength in Ovariectomized Rats. Front Pharmacol 2017. [PMID: 28638344 PMCID: PMC5461254 DOI: 10.3389/fphar.2017.00355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Osteoporosis is a major age-related bone disease characterized by low bone mineral density and a high risk of fractures. Bisphosphonates are considered as effective agents treating osteoporosis. However, long-term use of bisphosphonates is associated with some serious side effects, which limits the widespread clinical use of bisphosphonates. Here, we demonstrate a novel type of bone-targeting anti-resorptive agent, bisphosphonate-enoxacin (BE). In this study, ovariectomized rat model was established and treated with PBS, zoledronate (50 μg/kg) and different dose of BE (5 mg/kg and 10 mg/kg), respectively. The rats subjected to sham-operation and PBS treatment were considered as control group. Then, micro-computed tomography scanning, biomechanical tests, nano-indentation test and Raman analysis were used to compare the effects of zoledronate and BE on cortical bone mass, strength, and composition in ovariectomized rats. We found that both zoledronate and BE were beneficial to cortical bone strength. Three-point bending and nano-indentation tests showed that zoledronate- and BE-treated groups had superior general and local biomechanical properties compared to the ovariectomized groups. Interestingly, it seemed that BE-treated group got a better biomechanical property than the zoledronate-treated group. Also, BE-treated group showed significantly increased proteoglycan content compared with the zoledronate-treated group. We hypothesized that the increased bone strength and biomechanical properties was due to altered bone composition after treatment with BE. BE, a new bone-targeting agent, may be considered a more suitable anti-resorptive agent to treat osteoporosis and other bone diseases associated with decreased bone mass.
Collapse
Affiliation(s)
- Xuqiang Liu
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, The Artificial Joint Engineering and Technology Research Center of Jiangxi ProvinceNanchang, China
| | - Xinhua Qu
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Tao Nie
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, The Artificial Joint Engineering and Technology Research Center of Jiangxi ProvinceNanchang, China
| | - Zanjing Zhai
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Haowei Li
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Zhengxiao Ouyang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China.,Department of Orthopedics, The Second Xiangya Hospital, Central South UniversityChangsha, China
| | - An Qin
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Shuhong Zhang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Shuangyan Zhang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Qiming Fan
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Tingting Tang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Zhifeng Yu
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, The Artificial Joint Engineering and Technology Research Center of Jiangxi ProvinceNanchang, China
| |
Collapse
|
41
|
Goto Y, Aoyama M, Sekiya T, Kakita H, Waguri-Nagaya Y, Miyazawa K, Asai K, Goto S. CXCR4 + CD45 - Cells are Niche Forming for Osteoclastogenesis via the SDF-1, CXCL7, and CX3CL1 Signaling Pathways in Bone Marrow. Stem Cells 2016; 34:2733-2743. [PMID: 27339271 DOI: 10.1002/stem.2440] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/19/2016] [Accepted: 05/30/2016] [Indexed: 01/16/2023]
Abstract
Bone homeostasis comprises the balance between bone-forming osteoblasts and bone-resorbing osteoclasts (OCs), with an acceleration of osteoclastic bone resorption leading to osteoporosis. OCs can be generated from bone marrow cells (BMCs) under the tightly regulated local bone environment. However, it remained difficult to identify the critical cells responsible for providing an osteoclastogenesis niche. In this study, we used a fluorescence-activated cell sorting technique to determine the cell populations important for forming an appropriate microenvironment for osteoclastogenesis and to verify the associated interactions between osteoclast precursor cells and non-OCs. We isolated and removed a small cell population specific for osteoclastogenesis (CXCR4+ CD45- ) from mouse BMCs and cultured the remaining cells with receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage-colony stimulating factor. The resulting cultures showed significantly less large osteoclast formation. Quantitative RT-PCR analysis revealed that these CXCR4+ CD45- cells expressed low levels of RANK and RANKL, but high levels of critical chemokines including stromal cell derived factor 1 (SDF-1), chemokine (C-X-C motif) ligand 7 (CXCL7), and chemokine (C-X3-C motif) ligand 1 (CX3CL1). Furthermore, an SDF-1-specific antibody strongly suppressed OC formation in RAW264.7 cells and antibodies against SDF-1, CXCL7, and CX3CL1 suppressed OC formation in BMCs. These results suggest that isolated CXCR4+ CD45- cells support an appropriate microenvironment for osteoclastogenesis with a direct effect on the cells expressing SDF-1, CXCL7, and CX3CL1 receptors. The regulation of CXCR4+ CD45- cell function might therefore inform therapeutic strategies for diseases involving loss of bone homeostasis. Stem Cells 2016;34:2733-2743.
Collapse
Affiliation(s)
- Yoh Goto
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan.,Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mineyoshi Aoyama
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan
| | - Takeo Sekiya
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan.,Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroki Kakita
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Perinatal and Neonatal Medicine, Aichi Medical University, Nagakute, Japan
| | - Yuko Waguri-Nagaya
- Department of Joint Surgery for Rheumatic Diseases, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ken Miyazawa
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | - Kiyofumi Asai
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shigemi Goto
- Department of Orthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| |
Collapse
|
42
|
Thirukonda GJ, Uehara S, Nakayama T, Yamashita T, Nakamura Y, Mizoguchi T, Takahashi N, Yagami K, Udagawa N, Kobayashi Y. The dynamin inhibitor dynasore inhibits bone resorption by rapidly disrupting actin rings of osteoclasts. J Bone Miner Metab 2016; 34:395-405. [PMID: 26063501 DOI: 10.1007/s00774-015-0683-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/09/2015] [Indexed: 01/02/2023]
Abstract
The cytoskeletal organization of osteoclasts is required for bone resorption. Binding of dynamin with guanosine triphosphate (GTP) was previously suggested to be required for the organization of the actin cytoskeleton. However, the role of the GTPase activity of dynamin in the organization of the actin cytoskeleton as well as in the bone-resorbing activity of osteoclasts remains unclear. This study investigated the effects of dynasore, an inhibitor of the GTPase activity of dynamin, on the bone-resorbing activity of and actin ring formation in mouse osteoclasts in vitro and in vivo. Dynasore inhibited the formation of resorption pits in osteoclast cultures by suppressing actin ring formation and rapidly disrupting actin rings in osteoclasts. A time-lapse image analysis showed that dynasore shrank actin rings in osteoclasts within 30 min. The intraperitoneal administration of dynasore inhibited receptor activator of nuclear factor κB ligand (RANKL)-induced trabecular bone loss in mouse femurs. These in vitro and in vivo results suggest that the GTPase activity of dynamin is critical for the bone-resorbing activity of osteoclasts and that dynasore is a seed for the development of novel anti-resorbing agents.
Collapse
Affiliation(s)
- Gnanasagar J Thirukonda
- Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri-shi, Nagano, 399-0781, Japan
| | - Shunsuke Uehara
- Department of Biochemistry, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Takahiro Nakayama
- Department of Periodontology, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Teruhito Yamashita
- Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri-shi, Nagano, 399-0781, Japan
| | - Yukio Nakamura
- Department of Orthopaedic Surgery, School of Medicine, Shinshu University, Nagano, 390-8621, Japan
| | - Toshihide Mizoguchi
- Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri-shi, Nagano, 399-0781, Japan
| | - Naoyuki Takahashi
- Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri-shi, Nagano, 399-0781, Japan
| | - Kimitoshi Yagami
- Department of Oral Implantology, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Yasuhiro Kobayashi
- Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri-shi, Nagano, 399-0781, Japan.
| |
Collapse
|
43
|
Weir ME, Mann JE, Corwin T, Fulton ZW, Hao JM, Maniscalco JF, Kenney MC, Roman Roque KM, Chapdelaine EF, Stelzl U, Deming PB, Ballif BA, Hinkle KL. Novel autophosphorylation sites of Src family kinases regulate kinase activity and SH2 domain-binding capacity. FEBS Lett 2016; 590:1042-52. [PMID: 27001024 DOI: 10.1002/1873-3468.12144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/18/2016] [Accepted: 03/17/2016] [Indexed: 01/10/2023]
Abstract
Src family tyrosine kinases (SFKs) are critical players in normal and aberrant biological processes. While phosphorylation importantly regulates SFKs at two known tyrosines, large-scale phosphoproteomics have revealed four additional tyrosines commonly phosphorylated in SFKs. We found these novel tyrosines to be autophosphorylation sites. Mimicking phosphorylation at the C-terminal site to the activation loop decreased Fyn activity. Phosphomimetics and direct phosphorylation at the three SH2 domain sites increased Fyn activity while reducing phosphotyrosine-dependent interactions. While 68% of human SH2 domains exhibit conservation of at least one of these tyrosines, few have been found phosphorylated except when found in cis to a kinase domain.
Collapse
Affiliation(s)
- Marion E Weir
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Jacqueline E Mann
- Department of Medical Laboratory and Radiation Sciences, University of Vermont, Burlington, VT, USA
| | - Thomas Corwin
- Otto-Warburg Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Zachary W Fulton
- Department of Biology, University of Vermont, Burlington, VT, USA.,Department of Biology and Physical Education, Norwich University, Northfield, VT, USA
| | - Jennifer M Hao
- Department of Biology, University of Vermont, Burlington, VT, USA
| | | | - Marie C Kenney
- Department of Biology, University of Vermont, Burlington, VT, USA
| | | | - Elizabeth F Chapdelaine
- Department of Biology, University of Vermont, Burlington, VT, USA.,Department of Biology and Physical Education, Norwich University, Northfield, VT, USA
| | - Ulrich Stelzl
- Otto-Warburg Laboratory, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Paula B Deming
- Department of Medical Laboratory and Radiation Sciences, University of Vermont, Burlington, VT, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Karen L Hinkle
- Department of Biology, University of Vermont, Burlington, VT, USA.,Department of Biology and Physical Education, Norwich University, Northfield, VT, USA
| |
Collapse
|
44
|
Myristoleic acid inhibits osteoclast formation and bone resorption by suppressing the RANKL activation of Src and Pyk2. Eur J Pharmacol 2015; 768:189-98. [PMID: 26528796 DOI: 10.1016/j.ejphar.2015.10.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/29/2015] [Accepted: 10/30/2015] [Indexed: 02/01/2023]
Abstract
Cytoskeletal changes in osteoclasts such as formation of actin ring is required for bone-resorbing activity. The tyrosine kinase Src is a key player in massive cytoskeletal change of osteoclasts, thereby in bone destruction. In order for Src to be activated, trafficking to the inner plasma membrane via myristoylation is of importance. A previous study reported that myristoleic acid derived from myristic acid, inhibited N-myristoyl-transferase, an essential enzyme for myristoylation process. This prompted us to investigate whether myristoleic acid could affect osteoclastogenesis. Indeed, we observed that myristoleic acid inhibited RANKL-induced osteoclast formation in vitro, especially, at later stages of differentiation. Myristoleic acid attenuated the tyrosine phosphorylation of c-Src and Pyk2, which associates with Src, by RANKL. When myristoleic acid was co-administered with soluble RANKL into mice, RANKL-induced bone loss was substantially prevented. Bone dissection clearly revealed that the number of multinucleated osteoclasts was significantly diminished by myristoleic acid. On the other hand, myristoleic acid treatment had little or no influence on early osteoclast differentiation markers, such as c-Fos and NFATc1, and proteins related to cytoskeletal rearrangement, including DC-STAMP, integrin αv and integrin β3 in vitro. Taken together, our data suggest that myristoleic acid is capable of blocking the formation of large multinucleated osteoclasts and bone resorption likely through suppressing activation of Src and Pyk2.
Collapse
|
45
|
ten Harkel B, Schoenmaker T, Picavet DI, Davison NL, de Vries TJ, Everts V. The Foreign Body Giant Cell Cannot Resorb Bone, But Dissolves Hydroxyapatite Like Osteoclasts. PLoS One 2015; 10:e0139564. [PMID: 26426806 PMCID: PMC4591016 DOI: 10.1371/journal.pone.0139564] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/15/2015] [Indexed: 12/22/2022] Open
Abstract
Foreign body multinucleated giant cells (FBGCs) and osteoclasts share several characteristics, like a common myeloid precursor cell, multinuclearity, expression of tartrate-resistant acid phosphatase (TRAcP) and dendritic cell-specific transmembrane protein (DC-STAMP). However, there is an important difference: osteoclasts form and reside in the vicinity of bone, while FBGCs form only under pathological conditions or at the surface of foreign materials, like medical implants. Despite similarities, an important distinction between these cell types is that osteoclasts can resorb bone, but it is unknown whether FBGCs are capable of such an activity. To investigate this, we differentiated FBGCs and osteoclasts in vitro from their common CD14+ monocyte precursor cells, using different sets of cytokines. Both cell types were cultured on bovine bone slices and analyzed for typical osteoclast features, such as bone resorption, presence of actin rings, formation of a ruffled border, and characteristic gene expression over time. Additionally, both cell types were cultured on a biomimetic hydroxyapatite coating to discriminate between bone resorption and mineral dissolution independent of organic matrix proteolysis. Both cell types differentiated into multinucleated cells on bone, but FBGCs were larger and had a higher number of nuclei compared to osteoclasts. FBGCs were not able to resorb bone, yet they were able to dissolve the mineral fraction of bone at the surface. Remarkably, FBGCs also expressed actin rings, podosome belts and sealing zones--cytoskeletal organization that is considered to be osteoclast-specific. However, they did not form a ruffled border. At the gene expression level, FBGCs and osteoclasts expressed similar levels of mRNAs that are associated with the dissolution of mineral (e.g., anion exchange protein 2 (AE2), carbonic anhydrase 2 (CAII), chloride channel 7 (CIC7), and vacuolar-type H+-ATPase (v-ATPase)), in contrast the matrix degrading enzyme cathepsin K, which was hardly expressed by FBGCs. Functionally, the latter cells were able to dissolve a biomimetic hydroxyapatite coating in vitro, which was blocked by inhibiting v-ATPase enzyme activity. These results show that FBGCs have the capacity to dissolve the mineral phase of bone, similar to osteoclasts. However, they are not able to digest the matrix fraction of bone, likely due to the lack of a ruffled border and cathepsin K.
Collapse
Affiliation(s)
- Bas ten Harkel
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Daisy I. Picavet
- Department of Cell Biology and Histology, Center for Advanced Microscopy, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Noel L. Davison
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Xpand Biotechnology BV, Bilthoven, The Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute, University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Georgess D, Machuca-Gayet I, Blangy A, Jurdic P. Podosome organization drives osteoclast-mediated bone resorption. Cell Adh Migr 2015; 8:191-204. [PMID: 24714644 DOI: 10.4161/cam.27840] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Osteoclasts are the cells responsible for physiological bone resorption. A specific organization of their most prominent cytoskeletal structures, podosomes, is crucial for the degradation of mineralized bone matrix. Each podosome is constituted of an F-actin-enriched central core surrounded by a loose F-actin network, called the podosome cloud. In addition to intrinsic actin dynamics, podosomes are defined by their adhesion to the extracellular matrix, mainly via core-linking CD44 and cloud-linking integrins. These properties allow podosomes to collectively evolve into different patterns implicated in migration and bone resorption. Indeed, to resorb bone, osteoclasts polarize, actively secrete protons, and proteases into the resorption pit where these molecules are confined by a podosome-containing sealing zone. Here, we review recent advancements on podosome structure and regulatory pathways in osteoclasts. We also discuss the distinct functions of different podosome patterns during the lifespan of a single osteoclast.
Collapse
Affiliation(s)
- Dan Georgess
- Institut de Génomique Fonctionnelle de Lyon; Université de Lyon; Ecole Normale Supérieure de Lyon; Lyon, France
| | - Irma Machuca-Gayet
- Institut de Génomique Fonctionnelle de Lyon; Université de Lyon; Ecole Normale Supérieure de Lyon; Lyon, France
| | - Anne Blangy
- Centre de Recherche de Biochimie Macromoléculaire; CNRS UMR 5237; Montpellier University; Montpellier, France
| | - Pierre Jurdic
- Institut de Génomique Fonctionnelle de Lyon; Université de Lyon; Ecole Normale Supérieure de Lyon; Lyon, France
| |
Collapse
|
47
|
Choi YH, Han Y, Lee SH, Jin YH, Bahn M, Hur KC, Yeo CY, Lee KY. Cbl-b and c-Cbl negatively regulate osteoblast differentiation by enhancing ubiquitination and degradation of Osterix. Bone 2015; 75:201-9. [PMID: 25744063 DOI: 10.1016/j.bone.2015.02.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 11/28/2022]
Abstract
E3 ubiquitin ligase Cbl-b and c-Cbl play important roles in bone formation and maintenance. Cbl-b and c-Cbl regulate the activity of various receptor tyrosine kinases and intracellular protein tyrosine kinases mainly by regulating the degradation of target proteins. However, the precise mechanisms of how Cbl-b and c-Cbl regulate osteoblast differentiation are not well known. In this study, we investigated potential targets of Cbl-b and c-Cbl. We found that Cbl-b and c-Cbl inhibit BMP2-induced osteoblast differentiation in mesenchymal cells. Among various osteogenic transcription factors, we identified that Cbl-b and c-Cbl suppress the protein stability and transcriptional activity of Osterix. Our results suggest that Cbl-b and c-Cbl inhibit the function of Osterix by enhancing the ubiquitin-proteasome-mediated degradation of Osterix. Taken together, we propose novel regulatory roles of Cbl-b and c-Cbl during osteoblast differentiation in which Cbl-b and c-Cbl regulate the degradation of Osterix through the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- You Hee Choi
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Younho Han
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Sung Ho Lee
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Yun-Hye Jin
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Minjin Bahn
- Department of Life Science and Global Top5 Research Program, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Kyu Chung Hur
- Department of Life Science and Global Top5 Research Program, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Chang-Yeol Yeo
- Department of Life Science and Global Top5 Research Program, Ewha Womans University, Seoul 120-750, Republic of Korea.
| | - Kwang Youl Lee
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea.
| |
Collapse
|
48
|
Abstract
Knowledge of the molecular events that contribute to prostate cancer progression has created opportunities to develop novel therapy strategies. It is now well established that c-Src, a non-receptor tyrosine kinase, regulates a complex signaling network that drives the development of castrate-resistance and bone metastases, events that signal the lethal phenotype of advanced disease. Preclinical studies have established a role for c-Src and Src Family Kinases (SFKs) in proliferation, angiogenesis, invasion and bone metabolism, thus implicating Src signaling in both epithelial and stromal mechanisms of disease progression. A number of small molecule inhibitors of SFK now exist, many of which have demonstrated efficacy in preclinical models and several that have been tested in patients with metastatic castrate-resistant prostate cancer. These agents have demonstrated provocative clinic activity, particularly in modulating the bone microenvironment in a therapeutically favorable manner. Here, we review the discovery and basic biology of c-Src and further discuss the role of SFK inhibitors in the treatment of advanced prostate cancer.
Collapse
|
49
|
Adapala NS, Barbe MF, Tsygankov AY, Lorenzo JA, Sanjay A. Loss of Cbl-PI3K interaction enhances osteoclast survival due to p21-Ras mediated PI3K activation independent of Cbl-b. J Cell Biochem 2015; 115:1277-89. [PMID: 24470255 DOI: 10.1002/jcb.24779] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/24/2014] [Indexed: 01/14/2023]
Abstract
Cbl family proteins, Cbl and Cbl-b, are E3 ubiquitin ligases and adaptor proteins, which play important roles in bone-resorbing osteoclasts. Loss of Cbl in mice decreases osteoclast migration, resulting in delayed bone development where as absence of Cbl-b decreases bone volume due to hyper-resorptive osteoclasts. A major structural difference between Cbl and Cbl-b is tyrosine 737 (in YEAM motif) only on Cbl, which upon phosphorylation interacts with the p85 subunit of phosphatidylinositol-3 Kinase (PI3K). In contrast to Cbl(-/-) and Cbl-b(-/-) , mice lacking Cbl-PI3K interaction due to a Y737F (tyrosine to phenylalanine, YF) mutation showed enhanced osteoclast survival, but defective bone resorption. To investigate whether Cbl-PI3K interaction contributes to distinct roles of Cbl and Cbl-b in osteoclasts, mice bearing CblY737F mutation in the Cbl-b(-/-) background (YF/YF;Cbl-b(-/-) ) were generated. The differentiation and survival were augmented similarly in YF/YF and YF/YF;Cbl-b(-/-) osteoclasts, associated with enhanced PI3K signaling suggesting an exclusive role of Cbl-PI3K interaction, independent of Cbl-b. In addition to PI3K, the small GTPase Ras also regulates osteoclast survival. In the absence of Cbl-PI3K interaction, increased Ras GTPase activity and Ras-PI3K binding were observed and inhibition of Ras activation attenuated PI3K mediated osteoclast survival. In contrast to differentiation and survival, increased osteoclast activity observed in Cbl-b(-/-) mice persisted even after introduction of the resorption-defective YF mutation in YF/YF;Cbl-b(-/-) mice. Hence, Cbl and Cbl-b play mutually exclusive roles in osteoclasts. Whereas Cbl-PI3K interaction regulates differentiation and survival, bone resorption is predominantly regulated by Cbl-b in osteoclasts.
Collapse
Affiliation(s)
- Naga Suresh Adapala
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, 06032
| | | | | | | | | |
Collapse
|
50
|
Choi YH, Kim H, Han Y, Jin YH, Lee KY. Cbl regulates the activity of SIRT2. Biochem Biophys Res Commun 2014; 453:557-62. [DOI: 10.1016/j.bbrc.2014.09.125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 09/27/2014] [Indexed: 01/22/2023]
|