1
|
Sims NA. Osteoclast-derived coupling factors: origins and state-of-play Louis V Avioli lecture, ASBMR 2023. J Bone Miner Res 2024; 39:1377-1385. [PMID: 38990205 PMCID: PMC11425696 DOI: 10.1093/jbmr/zjae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Coupling, the mechanism that controls the sequence of events in bone remodeling, is a fundamental theory for understanding the way the skeleton changes throughout life. This review is an adapted version of the Louis V Avioli lecture, delivered at the Annual Scientific Meeting of the American Society of Bone and Mineral Research in 2023. It outlines the history of the coupling concept, details how coupling is thought to occur within trabecular and cortical bone, and describes its multiple contexts and the many mechanisms suggested to couple bone-forming osteoblasts to the prior action of osteoclasts on the same bone surface. These mechanisms include signals produced at each stage of the remodeling sequence (resorption, reversal, and formation), such as factors released by osteoclasts through their resorptive action and through protein synthesis, molecules deposited in the cement line during the reversal phase, and potential signals from osteocytes within the local bone environment. The review highlights two examples of coupling factors (Cardiotrophin 1 and EphrinB2:EphB4) to illustrate the limited data available, the need to integrate the many functions of these factors within the basic multicellular unit (BMU), and the multiple origins of these factors, including the other cell types present during the remodeling sequence (such as osteocytes, macrophages, endothelial cells, and T-cells).
Collapse
Affiliation(s)
- Natalie A Sims
- Bone Cell Biology and Diease Unit, St. Vincent’s Institute of Medical Research, Fitzroy, Victoria 3065, Australia
- Department of Medicine at St. Vincent’s Hospital Melbourne, The University of Melbourne, Fitzroy, Victoria 3065, Australia
- The Mary McKillop Institute for Health Research, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
2
|
Koh NYY, Miszkiewicz JJ, Fac ML, Wee NKY, Sims NA. Preclinical Rodent Models for Human Bone Disease, Including a Focus on Cortical Bone. Endocr Rev 2024; 45:493-520. [PMID: 38315213 PMCID: PMC11244217 DOI: 10.1210/endrev/bnae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/22/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Preclinical models (typically ovariectomized rats and genetically altered mice) have underpinned much of what we know about skeletal biology. They have been pivotal for developing therapies for osteoporosis and monogenic skeletal conditions, including osteogenesis imperfecta, achondroplasia, hypophosphatasia, and craniodysplasias. Further therapeutic advances, particularly to improve cortical strength, require improved understanding and more rigorous use and reporting. We describe here how trabecular and cortical bone structure develop, are maintained, and degenerate with aging in mice, rats, and humans, and how cortical bone structure is changed in some preclinical models of endocrine conditions (eg, postmenopausal osteoporosis, chronic kidney disease, hyperparathyroidism, diabetes). We provide examples of preclinical models used to identify and test current therapies for osteoporosis, and discuss common concerns raised when comparing rodent preclinical models to the human skeleton. We focus especially on cortical bone, because it differs between small and larger mammals in its organizational structure. We discuss mechanisms common to mouse and human controlling cortical bone strength and structure, including recent examples revealing genetic contributors to cortical porosity and osteocyte network configurations during growth, maturity, and aging. We conclude with guidelines for clear reporting on mouse models with a goal for better consistency in the use and interpretation of these models.
Collapse
Affiliation(s)
- Natalie Y Y Koh
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Justyna J Miszkiewicz
- School of Social Science, The University of Queensland, Brisbane, QLD 4072, Australia
- Vertebrate Evolution Development and Ecology, Naturalis Biodiversity Center, 2333 CR Leiden, The Netherlands
| | - Mary Louise Fac
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie K Y Wee
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie A Sims
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
3
|
Feng J, Huang Z, Lu J, Chan L, Feng X, Lei L, Huang Z, Lin L, Yao Y, Zhang X. Loss of signal transducer and activator of transcription 3 in osteoblasts impaired the bone healing in inflammatory microenvironment. Mol Oral Microbiol 2024; 39:136-151. [PMID: 37347649 DOI: 10.1111/omi.12425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/24/2023]
Abstract
INTRODUCTION This study aimed to investigate the effect of Stat3 on the osteoblast-mediated bone healing in the inflammatory lesion. METHODS The conditional knockout of Stat3 in osteoblasts (Stat3 CKO) was generated via the Cre-loxP recombination system using Osterix-Cre transgenic mice. The calvarial bone inflammatory lesions were established on both Stat3 CKO and wild-type mice, then harvested to assess the bone healing. In response to lipopolysaccharide (LPS) stimulation, osteoblasts from Stat3 CKO and wild-type mice were subjected to examine the formation of calcium deposits, the expression of osteogenic markers (i.e., Runx2, OPN, COL1A1), and osteoclast-related markers (i.e., RANKL, OPG). The EdU and transwell assays were performed to assess the proliferation and migration of the cells. RESULTS A decrease in bone mass and an increase in osteolysis were found in the inflammatory lesions on Stat3 CKO mice when compared with the control. More osteoclastic-like cells and an increased expression of RANKL were observed in Stat3 CKO mice. Both mRNA and protein expressions of Stat3 and osteogenic markers in the lesions were significantly decreased in Stat3 CKO mice. After co-cultured with osteogenic medium, the Stat3-deficient osteoblasts were found with a significant decrease in calcium deposits and the expression of osteogenic markers, and with a significant increased expression of RANKL. The impaired ossification of Stat3-deficient osteoblasts was even more pronounced with the presence of lipopolysaccharides in vitro. The most decrease in cell proliferation and migration was found in Stat3-deficient osteoblasts in response to LPS. CONCLUSIONS Loss of Stat3 in osteoblasts impaired bone healing in an inflammatory microenvironment.
Collapse
Affiliation(s)
- Jingyi Feng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Zijing Huang
- Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Jiarui Lu
- Department of Stomatology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| | - Laiting Chan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Xin Feng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Lizhen Lei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Zhuwei Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Lichieh Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Yichen Yao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
- Guangdong Province Key Laboratory of Stomatology, Guangzhou, Guangdong, P. R. China
| | - Xiaolei Zhang
- Department of Stomatology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, P. R. China
| |
Collapse
|
4
|
Sass JO, Sellin ML, Kauertz E, Johannsen J, Weinmann M, Stenzel M, Frank M, Vogel D, Bader R, Jonitz-Heincke A. Advanced Ti-Nb-Ta Alloys for Bone Implants with Improved Functionality. J Funct Biomater 2024; 15:46. [PMID: 38391899 PMCID: PMC10889793 DOI: 10.3390/jfb15020046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
The additive manufacturing of titanium-niobium-tantalum alloys with nominal chemical compositions Ti-xNb-6Ta (x = 20, 27, 35) by means of laser beam powder bed fusion is reported, and their potential as implant materials is elaborated by mechanical and biological characterization. The properties of dense specimens manufactured in different build orientations and of open porous Ti-20Nb-6Ta specimens are evaluated. Compression tests indicate that strength and elasticity are influenced by the chemical composition and build orientation. The minimum elasticity is always observed in the 90° orientation. It is lowest for Ti-20Nb-6Ta (43.2 ± 2.7 GPa) and can be further reduced to 8.1 ± 1.0 GPa for open porous specimens (p < 0.001). Furthermore, human osteoblasts are cultivated for 7 and 14 days on as-printed specimens and their biological response is compared to that of Ti-6Al-4V. Build orientation and cultivation time significantly affect the gene expression profile of osteogenic differentiation markers. Incomplete cell spreading is observed in specimens manufactured in 0° build orientation, whereas widely stretched cells are observed in 90° build orientation, i.e., parallel to the build direction. Compared to Ti-6Al-4V, Ti-Nb-Ta specimens promote improved osteogenesis and reduce the induction of inflammation. Accordingly, Ti-xNb-6Ta alloys have favorable mechanical and biological properties with great potential for application in orthopedic implants.
Collapse
Affiliation(s)
- Jan-Oliver Sass
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Marie-Luise Sellin
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Elisa Kauertz
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Jan Johannsen
- Fraunhofer Research Institution for Additive Manufacturing Technologies IAPT, Am Schleusengraben 14, 21029 Hamburg, Germany
| | | | | | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Strempelstraße 14, 18057 Rostock, Germany
- Department Life, Light and Matter, University of Rostock, 18051 Rostock, Germany
| | - Danny Vogel
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| | - Rainer Bader
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
- Department Life, Light and Matter, University of Rostock, 18051 Rostock, Germany
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Straße 142, 18057 Rostock, Germany
| |
Collapse
|
5
|
Yamamoto T, Abe M, Hongo H, Maruoka H, Yoshino H, Haraguchi-Kitakamae M, Udagawa N, Li M, Amizuka N, Hasegawa T. Differential osteoblastic activity in primary metaphyseal trabecular and secondary trabeculae of c-fos deficient mice. J Oral Biosci 2023; 65:265-272. [PMID: 37595744 DOI: 10.1016/j.job.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023]
Abstract
OBJECTIVES It has been highlighted that osteoblastic activities in remodeling-based bone formation are coupled with osteoclastic bone resorption while those in modeling-based bone formation are independent of osteoclasts. This study aimed to verify whether modeling-based bone formation can occur in the absence of osteoclasts. METHODS We performed histochemical analyses on the bone of eight-week-old male wild-type and c-fos-/- mice. Histochemical analyses were conducted on primary trabeculae near the chondro-osseous junction (COJ), sites of modeling-based bone formation, and secondary trabeculae, sites of remodeling-based bone formation, in the femora and tibiae of mice. RESULTS Alkaline phosphatase (ALP) immunoreactivity, a marker of osteoblastic lineages, was observed in the metaphyseal trabeculae of wild-type mice, while ALP was scattered throughout the femora of c-fos-/- mice. PHOSPHO1, an enzyme involved in matrix vesicle-mediated mineralization, was predominantly detected in primary trabeculae and also within short lines of osteoblasts in secondary trabeculae of wild-type mice. In contrast, femora of c-fos-/- mice showed several patches of PHOSPHO1 positivity in the primary trabeculae, but there were hardly any patches of PHOSPHO1 in secondary trabeculae. Calcein labeling was consistently observed in primary trabeculae close to the COJ in both wild-type and c-fos-/- mice; however, calcein labeling in the secondary trabeculae was only detected in wild-type mice. Transmission electron microscopic examination demonstrated abundant rough endoplasmic reticulum in the osteoblasts in secondary trabeculae of wild-type mice, but not in those of c-fos-/- mice. CONCLUSIONS Osteoblastic activities at the sites of modeling-based bone formation may be maintained in the absence of osteoclasts.
Collapse
Affiliation(s)
- Tomomaya Yamamoto
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan; Northern Army Medical Unit, Camp Makomanai, Japan Ground Self-Defense Forces, Sapporo, Japan
| | - Miki Abe
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Hiromi Hongo
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Haruhi Maruoka
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Hirona Yoshino
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Mai Haraguchi-Kitakamae
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan; Division of Craniofacial Development and Tissue Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Nobuyuki Udagawa
- Department of Oral Biochemistry, Matsumoto Dental University, Shiojiri, Japan
| | - Minqi Li
- Center of Osteoporosis and Bone Mineral Research, Department of Bone Metabolism, School of Stomatology, Shandong University, Jinan, China
| | - Norio Amizuka
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Tomoka Hasegawa
- Developmental Biology of Hard Tissue, Faculty of Dental Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
6
|
Li T, Cai K, Liu X, Cao H, Liu J, Wu L. Signal transducer and activator of transcription 3 positively regulates osteoblastic differentiation in MC3T3-E1 cells. Minerva Med 2023; 114:491-499. [PMID: 33047939 DOI: 10.23736/s0026-4806.20.06588-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Signal transducer and activator of transcription 3 (STAT3) plays a pivotal role in osteoblastic differentiation. However, the exact role of STAT3 in osteogenic differentiation of the pre-osteoblastic cell line MC3T3-E1 is still controversial. METHODS In this study, we demonstrated that eradication of STAT3 signaling by the inhibitors cryptotanshinone (CPT, a STAT3-specific inhibitor) or STAT3 siRNA both suppressed osteogenic differentiation of MC3T3-E1 cells, with a decrease in alkaline phosphatase (ALP) activity, protein expressions of the osteogenic differentiation markers Collagen I (ColI), ALP, and osteocalcin (OCN), and reduced matrix mineralization capacity at the terminal stage of osteogenic differentiation. However, the inhibition of STAT3 by CPT did not affect MC3T3-E1 cell proliferation. To further clarify the effect of STAT3 on osteogenic differentiation of MC3T3-E1 cells, we forced STAT3 expression and found that this ameliorated osteogenic differentiation. RESULTS Thus, our results confirmed that STAT3 is a likely positive regulator of osteogenic differentiation in MC3T3-E1 cells. CONCLUSIONS These findings may provide a basis for the development of more efficient and controllable protocols for osteoblastic differentiation and facilitate their use in regenerative medicine. In addition, our results provide novel insights into the effect of the STAT3 antagonist CPT on modulation of osteogenesis.
Collapse
Affiliation(s)
- Tian Li
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Kunzhan Cai
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Xiaohan Liu
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Hongjuan Cao
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Jie Liu
- Center of Science Experiment, China Medical University, Shenyang, China
| | - Lin Wu
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, China -
| |
Collapse
|
7
|
Jensen NR, Kelly RR, Kelly KD, Khoo SK, Sidles SJ, LaRue AC. From Stem to Sternum: The Role of Shp2 in the Skeleton. Calcif Tissue Int 2023; 112:403-421. [PMID: 36422682 DOI: 10.1007/s00223-022-01042-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/05/2022] [Indexed: 11/25/2022]
Abstract
Src homology-2 domain-containing phosphatase 2 (SHP2) is a ubiquitously expressed phosphatase that is vital for skeletal development and maintenance of chondrocytes, osteoblasts, and osteoclasts. Study of SHP2 function in small animal models has led to insights in phenotypes observed in SHP2-mutant human disease, such as Noonan syndrome. In recent years, allosteric SHP2 inhibitors have been developed to specifically target the protein in neoplastic processes. These inhibitors are highly specific and have great potential for disease modulation in cancer and other pathologies, including bone disorders. In this review, we discuss the importance of SHP2 and related signaling pathways (e.g., Ras/MEK/ERK, JAK/STAT, PI3K/Akt) in skeletal development. We review rodent models of pathologic processes caused by germline mutations that activate SHP2 enzymatic activity, with a focus on the skeletal phenotype seen in these patients. Finally, we discuss SHP2 inhibitors in development and their potential for disease modulation in these genetic diseases, particularly as it relates to the skeleton.
Collapse
Affiliation(s)
- Nathaniel R Jensen
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Ryan R Kelly
- Ralph H. Johnson VA Health Care System, Research Service, Charleston, SC, USA
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Kirsten D Kelly
- Ralph H. Johnson VA Health Care System, Research Service, Charleston, SC, USA
| | - Stephanie K Khoo
- Ralph H. Johnson VA Health Care System, Research Service, Charleston, SC, USA
| | - Sara J Sidles
- Ralph H. Johnson VA Health Care System, Research Service, Charleston, SC, USA
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda C LaRue
- Ralph H. Johnson VA Health Care System, Research Service, Charleston, SC, USA.
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
8
|
Abstract
Bone marrow contains resident cellular components that are not only involved in bone maintenance but also regulate hematopoiesis and immune responses. The immune system and bone interact with each other, coined osteoimmunology. Hashimoto's thyroiditis (HT) is one of the most common chronic autoimmune diseases which is accompanied by lymphocytic infiltration. It shows elevating thyroid autoantibody levels at an early stage and progresses to thyroid dysfunction ultimately. Different effects exert on bone metabolism during different phases of HT. In this review, we summarized the mechanisms of the long-term effects of HT on bone and the relationship between thyroid autoimmunity and osteoimmunology. For patients with HT, the bone is affected not only by thyroid function and the value of TSH, but also by the setting of the autoimmune background. The autoimmune background implies a breakdown of the mechanisms that control self-reactive system, featuring abnormal immune activation and presence of autoantibodies. The etiology of thyroid autoimmunity and osteoimmunology is complex and involves a number of immune cells, cytokines and chemokines, which regulate the pathogenesis of HT and osteoporosis at the same time, and have potential to affect each other. In addition, vitamin D works as a potent immunomodulator to influence both thyroid immunity and osteoimmunology. We conclude that HT affects bone metabolism at least through endocrine and immune pathways.
Collapse
Affiliation(s)
- Jialu Wu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, 37 Guoxue Lane, 610041, Chengdu, P.R. China
| | - Hui Huang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, 37 Guoxue Lane, 610041, Chengdu, P.R. China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, 37 Guoxue Lane, 610041, Chengdu, P.R. China.
| |
Collapse
|
9
|
Zhang J, Ye C, Zhu Y, Wang J, Liu J. The Cell-Specific Role of SHP2 in Regulating Bone Homeostasis and Regeneration Niches. Int J Mol Sci 2023; 24:ijms24032202. [PMID: 36768520 PMCID: PMC9917188 DOI: 10.3390/ijms24032202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Src homology-2 containing protein tyrosine phosphatase (SHP2), encoded by PTPN11, has been proven to participate in bone-related diseases, such as Noonan syndrome (NS), metachondromatosis and osteoarthritis. However, the mechanisms of SHP2 in bone remodeling and homeostasis maintenance are complex and undemonstrated. The abnormal expression of SHP2 can influence the differentiation and maturation of osteoblasts, osteoclasts and chondrocytes. Meanwhile, SHP2 mutations can act on the immune system, vasculature and nervous system, which in turn affect bone development and remodeling. Signaling pathways regulated by SHP2, such as mitogen-activated protein kinase (MAPK), Indian hedgehog (IHH) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT), are also involved in the proliferation, differentiation and migration of bone functioning cells. This review summarizes the recent advances of SHP2 on osteogenesis-related cells and niche cells in the bone marrow microenvironment. The phenotypic features of SHP2 conditional knockout mice and underlying mechanisms are discussed. The prospective applications of the current agonists or inhibitors that target SHP2 in bone-related diseases are also described. Full clarification of the role of SHP2 in bone remodeling will shed new light on potential treatment for bone related diseases.
Collapse
Affiliation(s)
- Jie Zhang
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chengxinyue Ye
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yufan Zhu
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (J.W.); (J.L.)
| | - Jin Liu
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (J.W.); (J.L.)
| |
Collapse
|
10
|
Isojima T, Walker EC, Poulton IJ, McGregor NE, Wicks IP, Gooi JH, Martin TJ, Sims NA. G-CSF Receptor Deletion Amplifies Cortical Bone Dysfunction in Mice With STAT3 Hyperactivation in Osteocytes. J Bone Miner Res 2022; 37:1876-1890. [PMID: 35856245 DOI: 10.1002/jbmr.4654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/27/2022] [Accepted: 07/16/2022] [Indexed: 11/09/2022]
Abstract
Bone strength is determined by the structure and composition of its thickened outer shell (cortical bone), yet the mechanisms controlling cortical consolidation are poorly understood. Cortical bone maturation depends on SOCS3-mediated suppression of IL-6 cytokine-induced STAT3 phosphorylation in osteocytes, the cellular network embedded in bone matrix. Because SOCS3 also suppresses granulocyte-colony-stimulating factor receptor (G-CSFR) signaling, we here tested whether global G-CSFR (Csf3r) ablation altereed bone structure in male and female mice lacking SOCS3 in osteocytes, (Dmp1Cre :Socs3f/f mice). Dmp1Cre :Socs3f/f :Csf3r-/- mice were generated by crossing Dmp1Cre :Socs3f/f mice with Csf3r-/- mice. Although G-CSFR is not expressed in osteocytes, Csf3r deletion further delayed cortical consolidation in Dmp1Cre :Socs3f/f mice. Micro-CT images revealed extensive, highly porous low-density bone, with little true cortex in the diaphysis, even at 26 weeks of age; including more low-density bone and less high-density bone in Dmp1Cre :Socs3f/f :Csf3r-/- mice than controls. By histology, the area where cortical bone would normally be found contained immature compressed trabecular bone in Dmp1Cre :Socs3f/f :Csf3r-/- mice and greater than normal levels of intracortical osteoclasts, extensive new woven bone formation, and the presence of more intracortical blood vessels than the already high levels observed in Dmp1Cre :Socs3f/f controls. qRT-PCR of cortical bone from Dmp1Cre :Socs3f/f :Csf3r-/- mice also showed more than a doubling of mRNA levels for osteoclasts, osteoblasts, RANKL, and angiogenesis markers. The further delay in cortical bone maturation was associated with significantly more phospho-STAT1 and phospho-STAT3-positive osteocytes, and a threefold increase in STAT1 and STAT3 target gene mRNA levels, suggesting G-CSFR deletion further increases STAT signaling beyond that of Dmp1Cre :Socs3f/f bone. G-CSFR deficiency therefore promotes STAT1/3 signaling in osteocytes, and when SOCS3 negative feedback is absent, elevated local angiogenesis, bone resorption, and bone formation delays cortical bone consolidation. This points to a critical role of G-CSF in replacing condensed trabecular bone with lamellar bone during cortical bone formation. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tsuyoshi Isojima
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Emma C Walker
- St. Vincent's Institute of Medical Research, Fitzroy, Australia
| | | | | | - Ian P Wicks
- Walter and Eliza Hall Institute, Parkville, Australia
| | - Jonathan H Gooi
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - T John Martin
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Australia
| |
Collapse
|
11
|
Effects of the Interleukin-6 Receptor Blocker Sarilumab on Metabolic Activity and Differentiation Capacity of Primary Human Osteoblasts. Pharmaceutics 2022; 14:pharmaceutics14071390. [PMID: 35890286 PMCID: PMC9318132 DOI: 10.3390/pharmaceutics14071390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/01/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Interleukin (IL-) 6 is a key factor in the inflammatory processes of rheumatoid arthritis. Several biologic agents target the IL-6 signaling pathway, including sarilumab, a monoclonal antibody that blocks the IL-6 receptor and inhibits IL-6-mediated cis- and trans-signaling. A careful analysis of the IL-6 signaling blockade should consider not only inflammatory processes but also the regenerative functions of IL-6. The purpose of this study was to investigate whether inhibition of the IL-6 receptors affects differentiation of human primary osteoblasts (hOB). The effects of sarilumab on viability and the differentiation capacity in unstimulated osteoblasts as well as after stimulation with various IL-6 and sIL6-R concentrations were determined. Sarilumab treatment alone did not affect the differentiation or induction of inflammatory processes in hOB. However, the significant induction of alkaline phosphatase activity which was observed after exogenous IL-6/sIL-6R costimulation at the highest concentrations was reduced back to baseline levels by the addition of sarilumab. The IL-6 receptor blockade also decreased gene expression of mediators required for osteogenesis and bone matrix maintenance. Our results demonstrate that concomitant administration of the IL-6 receptor blocker sarilumab can inhibit IL-6/sIL-6R-induced osteogenic differentiation.
Collapse
|
12
|
OMMA T, YÜCEL Ç, SERTOĞLU E, FIRAT SN, ÇULHA C, ÖZGÜRTAŞ T. The role of IL-6 and osteoprotegerin in bone metabolism in patients with Graves' disease. Turk J Med Sci 2022; 52:338-345. [PMID: 36161625 PMCID: PMC10381196 DOI: 10.55730/1300-0144.5320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 04/14/2022] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Increased bone turnover is a hallmark of hyperthyroidism. The underlying factors of how thyroid hormones affect bone cells are still under the spotlight. Previous studies indicated serum osteoprotegerin (OPG), receptor activator of NF-kB ligand (RANKL), and interleukin-6 (IL-6) as mediators of the effect of thyroid hormones on bone metabolism. Ultimately, the present research aimed to examine the association of IL-6 with OPG and RANKL in patients with hyperthyroidism. METHODS We carried out this study with 39 newly diagnosed and untreated Graves' patients and 43 healthy controls. In addition to routine tests, we measured serum OPG, RANKL, and IL-6 levels. RESULTS Mean age and sex distribution were similar in both groups. The hyperthyroid group had significantly higher OPG (p = 0.002) and IL-6 (p < 0.001) levels, but RANKL levels were significantly lower in this group (p < 0.001). We found OPG not to correlate with free T4 and T3, while it had a moderate and negative correlation with thyrotropin (TSH) (r = -0.372, p = 0.001). IL-6 had no correlation with OPG but positively correlated with free T4 (r = 0.445, p < 0.001) and free T3 (r = 0.326, p = 0.035). It also negatively correlated with RANKL (r = -0.247, p = 0.033). DISCUSSION Maintaining skeletal development and integrity is partially regulated by a normal balance of thyroid hormones. We concluded that increases in serum OPG and IL-6 levels accompanied hyperthyroidism. However, excessive levels of the hormones might cause drops in serum RANKL levels. Our results suggested that OPG, RANKL, and IL-6 might be involved in the cross-talking among immunity, thyroid function, and bone metabolism in the case of hyperthyroidism.
Collapse
Affiliation(s)
- Tülay OMMA
- Department of Endocrinology and Metabolism, Ankara Training and Research Hospital, University of Health Sciences, Ankara,
Turkey
| | - Çiğdem YÜCEL
- Department of Clinical Biochemistry, Ankara Numune Training and Research Hospital, University of Health Sciences, Ankara,
Turkey
| | - Erdim SERTOĞLU
- Department of Clinical Biochemistry, Ankara Gülhane Training and Research Hospital, University of Health Sciences, Ankara,
Turkey
| | - Sevde Nur FIRAT
- Department of Endocrinology and Metabolism, Ankara Training and Research Hospital, University of Health Sciences, Ankara,
Turkey
| | - Cavit ÇULHA
- Department of Endocrinology and Metabolism, Ankara Training and Research Hospital, University of Health Sciences, Ankara,
Turkey
| | - Taner ÖZGÜRTAŞ
- Department of Clinical Biochemistry, Ankara Gülhane Training and Research Hospital, University of Health Sciences, Ankara,
Turkey
| |
Collapse
|
13
|
de Souza PPC, Henning P, Lerner UH. Stimulation of Osteoclast Formation by Oncostatin M and the Role of WNT16 as a Negative Feedback Regulator. Int J Mol Sci 2022; 23:3287. [PMID: 35328707 PMCID: PMC8953253 DOI: 10.3390/ijms23063287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Oncostatin M (OSM), which belongs to the IL-6 family of cytokines, is the most potent and effective stimulator of osteoclast formation in this family, as assessed by different in vitro assays. Osteoclastogenesis induced by the IL-6 type of cytokines is mediated by the induction and paracrine stimulation of the osteoclastogenic cytokine receptor activator of nuclear factor κ-B ligand (RANKL), expressed on osteoblast cell membranes and targeting the receptor activator of nuclear factor κ-B (RANK) on osteoclast progenitor cells. The potent effect of OSM on osteoclastogenesis is due to an unusually robust induction of RANKL in osteoblasts through the OSM receptor (OSMR), mediated by a JAK-STAT/MAPK signaling pathway and by unique recruitment of the adapter protein Shc1 to the OSMR. Gene deletion of Osmr in mice results in decreased numbers of osteoclasts and enhanced trabecular bone caused by increased trabecular thickness, indicating that OSM may play a role in physiological regulation of bone remodeling. However, increased amounts of OSM, either through administration of recombinant protein or of adenoviral vectors expressing Osm, results in enhanced bone mass due to increased bone formation without any clear sign of increased osteoclast numbers, a finding which can be reconciled by cell culture experiments demonstrating that OSM can induce osteoblast differentiation and stimulate mineralization of bone nodules in such cultures. Thus, in vitro studies and gene deletion experiments show that OSM is a stimulator of osteoclast formation, whereas administration of OSM to mice shows that OSM is not a strong stimulator of osteoclastogenesis in vivo when administered to adult animals. These observations could be explained by our recent finding showing that OSM is a potent stimulator of the osteoclastogenesis inhibitor WNT16, acting in a negative feedback loop to reduce OSM-induced osteoclast formation.
Collapse
Affiliation(s)
- Pedro P. C. de Souza
- The Innovation in Biomaterials Laboratory, School of Dentistry, Federal University of Goiás, Goiânia 74690-900, Brazil;
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Ulf H. Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| |
Collapse
|
14
|
Protein tyrosine phosphatases in skeletal development and diseases. Bone Res 2022; 10:10. [PMID: 35091552 PMCID: PMC8799702 DOI: 10.1038/s41413-021-00181-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/29/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
Skeletal development and homeostasis in mammals are modulated by finely coordinated processes of migration, proliferation, differentiation, and death of skeletogenic cells originating from the mesoderm and neural crest. Numerous molecular mechanisms are involved in these regulatory processes, one of which is protein posttranslational modifications, particularly protein tyrosine phosphorylation (PYP). PYP occurs mainly through the action of protein tyrosine kinases (PTKs), modifying protein enzymatic activity, changing its cellular localization, and aiding in the assembly or disassembly of protein signaling complexes. Under physiological conditions, PYP is balanced by the coordinated action of PTKs and protein tyrosine phosphatases (PTPs). Dysregulation of PYP can cause genetic, metabolic, developmental, and oncogenic skeletal diseases. Although PYP is a reversible biochemical process, in contrast to PTKs, little is known about how this equilibrium is modulated by PTPs in the skeletal system. Whole-genome sequencing has revealed a large and diverse superfamily of PTP genes (over 100 members) in humans, which can be further divided into cysteine (Cys)-, aspartic acid (Asp)-, and histidine (His)-based PTPs. Here, we review current knowledge about the functions and regulatory mechanisms of 28 PTPs involved in skeletal development and diseases; 27 of them belong to class I and II Cys-based PTPs, and the other is an Asp-based PTP. Recent progress in analyzing animal models that harbor various mutations in these PTPs and future research directions are also discussed. Our literature review indicates that PTPs are as crucial as PTKs in supporting skeletal development and homeostasis.
Collapse
|
15
|
Liu NQ, Lin Y, Li L, Lu J, Geng D, Zhang J, Jashashvili T, Buser Z, Magallanes J, Tassey J, Shkhyan R, Sarkar A, Lopez N, Lee S, Lee Y, Wang L, Petrigliano FA, Van Handel B, Lyons K, Evseenko D. gp130/STAT3 signaling is required for homeostatic proliferation and anabolism in postnatal growth plate and articular chondrocytes. Commun Biol 2022; 5:64. [PMID: 35039652 PMCID: PMC8763901 DOI: 10.1038/s42003-021-02944-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023] Open
Abstract
Growth of long bones and vertebrae is maintained postnatally by a long-lasting pool of progenitor cells. Little is known about the molecular mechanisms that regulate the output and maintenance of the cells that give rise to mature cartilage. Here we demonstrate that postnatal chondrocyte-specific deletion of a transcription factor Stat3 results in severely reduced proliferation coupled with increased hypertrophy, growth plate fusion, stunting and signs of progressive dysfunction of the articular cartilage. This effect is dimorphic, with females more strongly affected than males. Chondrocyte-specific deletion of the IL-6 family cytokine receptor gp130, which activates Stat3, phenocopied Stat3-deletion; deletion of Lifr, one of many co-receptors that signals through gp130, resulted in a milder phenotype. These data define a molecular circuit that regulates chondrogenic cell maintenance and output and reveals a pivotal positive function of IL-6 family cytokines in the skeletal system with direct implications for skeletal development and regeneration.
Collapse
Affiliation(s)
- Nancy Q. Liu
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Yucheng Lin
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.263826.b0000 0004 1761 0489Department of Orthopaedic Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009 China
| | - Liangliang Li
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.89957.3a0000 0000 9255 8984Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu 211100 China
| | - Jinxiu Lu
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Dawei Geng
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Jiankang Zhang
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 China
| | - Tea Jashashvili
- grid.42505.360000 0001 2156 6853Department of Radiology, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Zorica Buser
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Jenny Magallanes
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Jade Tassey
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Ruzanna Shkhyan
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Arijita Sarkar
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Noah Lopez
- grid.19006.3e0000 0000 9632 6718Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angles (UCLA), Los Angeles, CA USA
| | - Siyoung Lee
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Youngjoo Lee
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Liming Wang
- grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.89957.3a0000 0000 9255 8984Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu 210006 China
| | - Frank A. Petrigliano
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA 90033 USA
| | - Ben Van Handel
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Karen Lyons
- grid.19006.3e0000 0000 9632 6718Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angles (UCLA), Los Angeles, CA USA
| | - Denis Evseenko
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA 90033 USA
| |
Collapse
|
16
|
Jenkins RH, Hughes STO, Figueras AC, Jones SA. Unravelling the broader complexity of IL-6 involvement in health and disease. Cytokine 2021; 148:155684. [PMID: 34411990 DOI: 10.1016/j.cyto.2021.155684] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
The classification of interleukin-6 (IL-6) as a pro-inflammatory cytokine undervalues the biological impact of this cytokine in health and disease. With broad activities affecting the immune system, tissue homeostasis and metabolic processes, IL-6 displays complex biology. The significance of these involvements has become increasingly important in clinical settings where IL-6 is identified as a prominent target for therapy. Here, clinical experience with IL-6 antagonists emphasises the need to understand the context-dependent properties of IL-6 within an inflammatory environment and the anticipated or unexpected consequences of IL-6 blockade. In this review, we will describe the immunobiology of IL-6 and explore the gamut of IL-6 bioactivity affecting the clinical response to biological drugs targeting this cytokine pathway.
Collapse
Affiliation(s)
- Robert H Jenkins
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Stuart T O Hughes
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Ana Cardus Figueras
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Simon A Jones
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
17
|
Influences of the IL-6 cytokine family on bone structure and function. Cytokine 2021; 146:155655. [PMID: 34332274 DOI: 10.1016/j.cyto.2021.155655] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023]
Abstract
The IL-6 family of cytokines comprises a large group of cytokines that all act via the formation of a signaling complex that includes the glycoprotein 130 (gp130) receptor. Despite this, many of these cytokines have unique roles that regulate the activity of bone forming osteoblasts, bone resorbing osteoclasts, bone-resident osteocytes, and cartilage cells (chondrocytes). These include specific functions in craniofacial development, longitudinal bone growth, and the maintenance of trabecular and cortical bone structure, and have been implicated in musculoskeletal pathologies such as craniosynostosis, osteoporosis, rheumatoid arthritis, osteoarthritis, and heterotopic ossifications. This review will work systematically through each member of this family and provide an overview and an update on the expression patterns and functions of each of these cytokines in the skeleton, as well as their negative feedback pathways, particularly suppressor of cytokine signaling 3 (SOCS3). The specific cytokines described are interleukin 6 (IL-6), interleukin 11 (IL-11), oncostatin M (OSM), leukemia inhibitory factor (LIF), cardiotrophin 1 (CT-1), ciliary neurotrophic factor (CNTF), cardiotrophin-like cytokine factor 1 (CLCF1), neuropoietin, humanin and interleukin 27 (IL-27).
Collapse
|
18
|
IL-6 regulates the bone metabolism and inflammatory microenvironment in aging mice by inhibiting Setd7. Acta Histochem 2021; 123:151718. [PMID: 33962150 DOI: 10.1016/j.acthis.2021.151718] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/21/2022]
Abstract
Aging, which has become a worldwide problem, leads to the degeneration of multiple organs and tissues. Two of the main changes in aging are dysregulation of the tissue microenvironment and abnormal functioning of specific stem cells. Bone marrow stem cells (BMSCs) in the aging microenvironment are not only effector cells but also immunomodulatory cells that change the microenvironment. IL-6 is a primary inflammatory response factor associated with bone diseases. In this study, we stimulated BMSCs with IL-6 to investigate a novel mechanism of age-related osteoporosis. IL-6 activated the TLR2, TLR4 and AKT pathway as well as inhibited the expression of β-catenin and Setd7. In addition, Setd7 expression in the bone tissues of aged mice was suppressed. Setd7 not only promoted BMSC osteogenic differentiation but also mediated proinflammatory gene expression in BMSCs under IL-6 stimulation. Due to its dual functions in BMSCs, Setd7 may be a novel molecular target for age-related osteoporosis prevention and treatment.
Collapse
|
19
|
Adam S, Simon N, Steffen U, Andes FT, Scholtysek C, Müller DIH, Weidner D, Andreev D, Kleyer A, Culemann S, Hahn M, Schett G, Krönke G, Frey S, Hueber AJ. JAK inhibition increases bone mass in steady-state conditions and ameliorates pathological bone loss by stimulating osteoblast function. Sci Transl Med 2021; 12:12/530/eaay4447. [PMID: 32051226 DOI: 10.1126/scitranslmed.aay4447] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/08/2020] [Indexed: 12/17/2022]
Abstract
Janus kinase (JAK)-mediated cytokine signaling has emerged as an important therapeutic target for the treatment of inflammatory diseases such as rheumatoid arthritis (RA). Accordingly, JAK inhibitors compose a new class of drugs, among which tofacitinib and baricitinib have been approved for the treatment of RA. Periarticular bone erosions contribute considerably to the pathogenesis of RA. However, although the immunomodulatory aspect of JAK inhibition (JAKi) is well defined, the current knowledge of how JAKi influences bone homeostasis is limited. Here, we assessed the effects of the JAK inhibitors tofacitinib and baricitinib on bone phenotype (i) in mice during steady-state conditions or in mice with bone loss induced by (ii) estrogen-deficiency (ovariectomy) or (iii) inflammation (arthritis) to evaluate whether effects of JAKi on bone metabolism require noninflammatory/inflammatory challenge. In all three models, JAKi increased bone mass, consistent with reducing the ratio of receptor activator of NF-κB ligand/osteoprotegerin in serum. In vitro, effects of tofacitinib and baricitinib on osteoclast and osteoblast differentiation were analyzed. JAKi significantly increased osteoblast function (P < 0.05) but showed no direct effects on osteoclasts. Additionally, mRNA sequencing and ingenuity pathway analyses were performed in osteoblasts exposed to JAKi and revealed robust up-regulation of markers for osteoblast function, such as osteocalcin and Wnt signaling. The anabolic effect of JAKi was illustrated by the stabilization of β-catenin. In humans with RA, JAKi induced bone-anabolic effects as evidenced by repair of arthritic bone erosions. Results support that JAKi is a potent therapeutic tool for increasing osteoblast function and bone formation.
Collapse
Affiliation(s)
- Susanne Adam
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Nils Simon
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Ulrike Steffen
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Fabian T Andes
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Carina Scholtysek
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Dorothea I H Müller
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Daniela Weidner
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Darja Andreev
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Arnd Kleyer
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Stephan Culemann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Madelaine Hahn
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Georg Schett
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Gerhard Krönke
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Silke Frey
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany. .,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Axel J Hueber
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
20
|
The downstream RAF-1 signaling of fibroblast growth factor-23 participates in the osteogenetic effect caused by C-type natriuretic peptide in vitro. Adv Med Sci 2021; 66:206-214. [PMID: 33735829 DOI: 10.1016/j.advms.2021.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/27/2020] [Accepted: 03/03/2021] [Indexed: 11/23/2022]
Abstract
PURPOSE Several studies have demonstrated that C-type natriuretic peptide (CNP) stimulates osteoblastic proliferation seemly via antagonizing the expression of fibroblast growth factor (FGF)-23 in vitro. The main aim of the present study is to probe whether the post-receptor pathways of FGF-23 participate in osteogenesis caused by CNP. METHODS Osteoblasts were cultured in the absence or presence of CNP: 0, 10, and 100 pmol/L, for 24 h, 48 h and 72 h, respectively. RESULTS The findings of the present study indicated that osteoblastic proliferation was directly promoted by exogenous CNP in a dose-dependent manner; osteoblastic FGF-23 was significantly down-regulated by CNP at 24 h post-treatment; RAF-1, extracellular signal-regulated kinases (ERK), and P38 were substantially suppressed by CNP in a dose- and time-dependent manner; and signal transducer and activator of transcription (STAT)-1 was not changed on the premise of the down-regulated FGF-23 in osteoblasts treated with CNP. CONCLUSION CNP may promote osteogenesis via inhibiting ERK and P38, rather than STAT-1, in the downstream of FGF-23/RAF-1 pathway.
Collapse
|
21
|
Pang L, Huynh J, Alorro MG, Li X, Ernst M, Chand AL. STAT3 Signalling via the IL-6ST/gp130 Cytokine Receptor Promotes Epithelial Integrity and Intestinal Barrier Function during DSS-Induced Colitis. Biomedicines 2021; 9:biomedicines9020187. [PMID: 33673239 PMCID: PMC7918037 DOI: 10.3390/biomedicines9020187] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/22/2022] Open
Abstract
The intestinal epithelium provides a barrier against commensal and pathogenic microorganisms. Barrier dysfunction promotes chronic inflammation, which can drive the pathogenesis of inflammatory bowel disease (IBD) and colorectal cancer (CRC). Although the Signal Transducer and Activator of Transcription-3 (STAT3) is overexpressed in both intestinal epithelial cells and immune cells in IBD patients, the role of the interleukin (IL)-6 family of cytokines through the shared IL-6ST/gp130 receptor and its associated STAT3 signalling in intestinal barrier integrity is unclear. We therefore investigated the role of STAT3 in retaining epithelial barrier integrity using dextran sulfate sodium (DSS)-induced colitis in two genetically modified mouse models, to either reduce STAT1/3 activation in response to IL-6 family cytokines with a truncated gp130∆STAT allele (GP130∆STAT/+), or by inducing short hairpin-mediated knockdown of Stat3 (shStat3). Here, we show that mice with reduced STAT3 activity are highly susceptible to DSS-induced colitis. Mechanistically, the IL-6/gp130/STAT3 signalling cascade orchestrates intestinal barrier function by modulating cytokine secretion and promoting epithelial integrity to maintain a defence against bacteria. Our study also identifies a crucial role of STAT3 in controlling intestinal permeability through tight junction proteins. Thus, therapeutically targeting the IL-6/gp130/STAT3 signalling axis to promote barrier function may serve as a treatment strategy for IBD patients.
Collapse
Affiliation(s)
- Lokman Pang
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; (J.H.); (M.G.A.); (M.E.)
- Correspondence: (L.P.); (A.L.C.)
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; (J.H.); (M.G.A.); (M.E.)
| | - Mariah G. Alorro
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; (J.H.); (M.G.A.); (M.E.)
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Bundoora, VIC 3083, Australia;
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; (J.H.); (M.G.A.); (M.E.)
| | - Ashwini L. Chand
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC 3084, Australia; (J.H.); (M.G.A.); (M.E.)
- Correspondence: (L.P.); (A.L.C.)
| |
Collapse
|
22
|
Abstract
Obesity and obesity-related diseases like type 2 diabetes (T2D) are prominent global health issues; therefore, there is a need to better understand the mechanisms underlying these conditions. The onset of obesity is characterized by accumulation of proinflammatory cells, including Ly6chi monocytes (which differentiate into proinflammatory macrophages) and neutrophils, in metabolic tissues. This shift toward chronic, low-grade inflammation is an obese-state hallmark and highly linked to metabolic disorders and other obesity comorbidities. The mechanisms that induce and maintain increased inflammatory myelopoiesis are of great interest, with a recent focus on how obesity affects more primitive hematopoietic cells. The hematopoietic system is constantly replenished by proper regulation of hematopoietic stem and progenitor (HSPC) pools in the BM. While early research suggests that chronic obesity promotes expansion of myeloid-skewed HSPCs, the involvement of the hematopoietic stem cell (HSC) niche in regulating obesity-induced myelopoiesis remains undefined. In this review, we explore the role of the multicellular HSC niche in hematopoiesis and inflammation, and the potential contribution of this niche to the hematopoietic response to obesity. This review further aims to summarize the potential HSC niche involvement as a target of obesity-induced inflammation and a driver of obesity-induced myelopoiesis.
Collapse
|
23
|
Estell EG, Rosen CJ. Emerging insights into the comparative effectiveness of anabolic therapies for osteoporosis. Nat Rev Endocrinol 2021; 17:31-46. [PMID: 33149262 DOI: 10.1038/s41574-020-00426-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 01/01/2023]
Abstract
Over the past three decades, the mainstay of treatment for osteoporosis has been antiresorptive agents (such as bisphosphonates), which have been effective with continued administration in lowering fracture risk. However, the clinical landscape has changed as adherence to these medications has declined due to perceived adverse effects. As a result, decreases in hip fracture rates that followed the introduction of bisphosphonates have now levelled off, which is coincident with a decline in the use of the antiresorptive agents. In the past two decades, two types of anabolic agents (including three new drugs), which represent a novel approach to improving bone quality by increasing bone formation, have been approved. These therapies are expected to lead to a new clinical paradigm in which anabolic agents will be used either alone or in combination with antiresorptive agents to build new bone and reduce fracture risk. This Review examines the mechanisms of action for these anabolic agents by detailing their receptor-activating properties for key cell types in the bone and marrow niches. Using these advances in bone biology as context, the comparative effectiveness of these anabolic agents is discussed in relation to other therapeutic options for osteoporosis to better guide their clinical application in the future.
Collapse
Affiliation(s)
- Eben G Estell
- Maine Medical Center Research Institute, Scarborough, ME, USA
| | | |
Collapse
|
24
|
Chen YH, Grigelioniene G, Newton PT, Gullander J, Elfving M, Hammarsjö A, Batkovskyte D, Alsaif HS, Kurdi WIY, Abdulwahab F, Shanmugasundaram V, Devey L, Bacrot S, Brodszki J, Huber C, Hamel B, Gisselsson D, Papadogiannakis N, Jedrycha K, Gürtl-Lackner B, Chagin AS, Nishimura G, Aschenbrenner D, Alkuraya FS, Laurence A, Cormier-Daire V, Uhlig HH. Absence of GP130 cytokine receptor signaling causes extended Stüve-Wiedemann syndrome. J Exp Med 2020; 217:133568. [PMID: 31914175 PMCID: PMC7062520 DOI: 10.1084/jem.20191306] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/07/2019] [Accepted: 11/14/2019] [Indexed: 01/25/2023] Open
Abstract
The gene IL6ST encodes GP130, the common signal transducer of the IL-6 cytokine family consisting of 10 cytokines. Previous studies have identified cytokine-selective IL6ST defects that preserve LIF signaling. We describe three unrelated families with at least five affected individuals who presented with lethal Stüve-Wiedemann–like syndrome characterized by skeletal dysplasia and neonatal lung dysfunction with additional features such as congenital thrombocytopenia, eczematoid dermatitis, renal abnormalities, and defective acute-phase response. We identified essential loss-of-function variants in IL6ST (a homozygous nonsense variant and a homozygous intronic splice variant with exon skipping). Functional tests showed absent cellular responses to GP130-dependent cytokines including IL-6, IL-11, IL-27, oncostatin M (OSM), and leukemia inhibitory factor (LIF). Genetic reconstitution of GP130 by lentiviral transduction in patient-derived cells reversed the signaling defect. This study identifies a new genetic syndrome caused by the complete lack of signaling of a whole family of GP130-dependent cytokines in humans and highlights the importance of the LIF signaling pathway in pre- and perinatal development.
Collapse
Affiliation(s)
- Yin-Huai Chen
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Giedre Grigelioniene
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Phillip T Newton
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jacob Gullander
- University and Regional Laboratories Department of Clinical Genetics, Lund, Sweden
| | - Maria Elfving
- Department of Clinical Sciences, Pediatrics, Skåne University Hospital Lund, Lund University, Lund, Sweden
| | - Anna Hammarsjö
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Dominyka Batkovskyte
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hessa S Alsaif
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Wesam I Y Kurdi
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Firdous Abdulwahab
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | | | - Séverine Bacrot
- Department of Clinical Genetics, INSERM UMR 1163, Université Paris Descartes-Sorbonne Paris cité, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| | - Jana Brodszki
- Department of Obstetrics and Gynecology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Celine Huber
- Department of Clinical Genetics, INSERM UMR 1163, Université Paris Descartes-Sorbonne Paris cité, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| | - Ben Hamel
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - David Gisselsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Sweden
| | - Nikos Papadogiannakis
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Katarina Jedrycha
- Department of Clinical Sciences, Pediatrics, Skåne University Hospital Lund, Lund University, Lund, Sweden
| | - Barbara Gürtl-Lackner
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Sweden
| | - Andrei S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russian Federation
| | - Gen Nishimura
- Center for Intractable Diseases, Saitama University Hospital, Saitama, Japan
| | | | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Arian Laurence
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Valérie Cormier-Daire
- Department of Clinical Genetics, INSERM UMR 1163, Université Paris Descartes-Sorbonne Paris cité, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.,Department of Paediatrics, University of Oxford, Oxford, UK.,Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK
| |
Collapse
|
25
|
Sims NA. The JAK1/STAT3/SOCS3 axis in bone development, physiology, and pathology. Exp Mol Med 2020; 52:1185-1197. [PMID: 32788655 PMCID: PMC8080635 DOI: 10.1038/s12276-020-0445-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Abstract
Bone growth and the maintenance of bone structure are controlled by multiple endocrine and paracrine factors, including cytokines expressed locally within the bone microenvironment and those that are elevated, both locally and systemically, under inflammatory conditions. This review focuses on those bone-active cytokines that initiate JAK–STAT signaling, and outlines the discoveries made from studying skeletal defects caused by induced or spontaneous modifications in this pathway. Specifically, this review describes defects in JAK1, STAT3, and SOCS3 signaling in mouse models and in humans, including mutations designed to modify these pathways downstream of the gp130 coreceptor. It is shown that osteoclast formation is generally stimulated indirectly by these pathways through JAK1 and STAT3 actions in inflammatory and other accessory cells, including osteoblasts. In addition, in bone remodeling, osteoblast differentiation is increased secondary to stimulated osteoclast formation through an IL-6-dependent pathway. In growth plate chondrocytes, STAT3 signaling promotes the normal differentiation process that leads to bone lengthening. Within the osteoblast lineage, STAT3 signaling promotes bone formation in normal physiology and in response to mechanical loading through direct signaling in osteocytes. This activity, particularly that of the IL-6/gp130 family of cytokines, must be suppressed by SOCS3 for the normal formation of cortical bone. Maintaining normal bone structure and strength depends on a group of signaling proteins called cytokines that bind to receptor molecules on cell surfaces. Natalie Sims at St. Vincent’s Institute of Medical Research and The University of Melbourne in Australia reviews the role of cytokines in a specific signaling pathway in bone development and disease. Two of the proteins in this pathway respond to cytokine activity, whereas the third inhibits the cytokines’ effects. Studies in mice and humans have identified links between specific bone defects and spontaneous or experimentally induced mutations in the genes that code for the three proteins. The review covers the significance of recent findings to several types of cells that form new bone, degrade bone as part of normal bone turnover, and sustain the structure of bone and cartilage.
Collapse
Affiliation(s)
- Natalie A Sims
- St. Vincent's Institute of Medical Research, and Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
26
|
Abstract
The skeleton is highly vascularized due to the various roles blood vessels play in the homeostasis of bone and marrow. For example, blood vessels provide nutrients, remove metabolic by-products, deliver systemic hormones, and circulate precursor cells to bone and marrow. In addition to these roles, bone blood vessels participate in a variety of other functions. This article provides an overview of the afferent, exchange and efferent vessels in bone and marrow and presents the morphological layout of these blood vessels regarding blood flow dynamics. In addition, this article discusses how bone blood vessels participate in bone development, maintenance, and repair. Further, mechanical loading-induced bone adaptation is presented regarding interstitial fluid flow and pressure, as regulated by the vascular system. The role of the sympathetic nervous system is discussed in relation to blood vessels and bone. Finally, vascular participation in bone accrual with intermittent parathyroid hormone administration, a medication prescribed to combat age-related bone loss, is described and age- and disease-related impairments in blood vessels are discussed in relation to bone and marrow dysfunction. © 2020 American Physiological Society. Compr Physiol 10:1009-1046, 2020.
Collapse
Affiliation(s)
- Rhonda D Prisby
- Bone Vascular and Microcirculation Laboratory, Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
27
|
Walker EC, Truong K, McGregor NE, Poulton IJ, Isojima T, Gooi JH, Martin TJ, Sims NA. Cortical bone maturation in mice requires SOCS3 suppression of gp130/STAT3 signalling in osteocytes. eLife 2020; 9:e56666. [PMID: 32458800 PMCID: PMC7253175 DOI: 10.7554/elife.56666] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/01/2020] [Indexed: 12/23/2022] Open
Abstract
Bone strength is determined by its dense cortical shell, generated by unknown mechanisms. Here we use the Dmp1Cre:Socs3f/f mouse, with delayed cortical bone consolidation, to characterise cortical maturation and identify control signals. We show that cortical maturation requires a reduction in cortical porosity, and a transition from low to high density bone, which continues even after cortical shape is established. Both processes were delayed in Dmp1Cre:Socs3f/f mice. SOCS3 (suppressor of cytokine signalling 3) inhibits signalling by leptin, G-CSF, and IL-6 family cytokines (gp130). In Dmp1Cre:Socs3f/f bone, STAT3 phosphorylation was prolonged in response to gp130-signalling cytokines, but not G-CSF or leptin. Deletion of gp130 in Dmp1Cre:Socs3f/f mice suppressed STAT3 phosphorylation in osteocytes and osteoclastic resorption within cortical bone, leading to rescue of the corticalisation defect, and restoration of compromised bone strength. We conclude that cortical bone development includes both pore closure and accumulation of high density bone, and that these processes require suppression of gp130-STAT3 signalling in osteocytes.
Collapse
Affiliation(s)
- Emma C Walker
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
| | - Kim Truong
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- University of Melbourne, Department of Medicine at St. Vincent’s HospitalFitzroyAustralia
| | | | | | - Tsuyoshi Isojima
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- Department of Pediatrics, Teikyo University School of MedicineTokyoJapan
| | - Jonathan H Gooi
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of MelbourneParkvilleAustralia
| | - T John Martin
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- University of Melbourne, Department of Medicine at St. Vincent’s HospitalFitzroyAustralia
| | - Natalie A Sims
- St. Vincent’s Institute of Medical ResearchFitzroyAustralia
- University of Melbourne, Department of Medicine at St. Vincent’s HospitalFitzroyAustralia
| |
Collapse
|
28
|
Functional Block of Interleukin-6 Reduces a Bone Pain Marker but Not Bone Loss in Hindlimb-Unloaded Mice. Int J Mol Sci 2020; 21:ijms21103521. [PMID: 32429268 PMCID: PMC7278999 DOI: 10.3390/ijms21103521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/08/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Interleukin-6 (IL-6) is widely accepted to stimulate osteoclasts. Our aim in this study was to examine whether the inhibitory effect of IL-6 on bone loss and skeletal pain associated with osteoporosis in hindlimb-unloaded (HU) mice in comparison with bisphosphonate. Eight-week-old male ddY mice were tail suspended for 2 weeks. Starting immediately after reload, vehicle (HU group), alendronate (HU-ALN group), or anti-IL-6 receptor antibody (HU-IL-6i group) was injected subcutaneously. After a 2-week treatment, pain-related behavior was examined using von Frey filaments. The bilateral distal femoral and proximal tibial metaphyses were analyzed three-dimensionally with micro-computed tomography. Calcitonin gene-related peptide (CGRP) expressions in dorsal root ganglion (DRG) neurons innervating the hindlimbs were examined using immunohistochemistry. HU mice with tail suspension developed bone loss. The HU mice showed mechanical hyperalgesia in the hindlimbs and increased CGRP immunoreactive neurons in the L3-5 DRG. Treatment with IL-6i and ALN prevented HU-induced mechanical hyperalgesia and upregulation of CGRP expressions in DRG neurons. Furthermore, ALN but not IL-6i prevented HU-induced bone loss. In summary, treatment with IL-6i prevented mechanical hyperalgesia in hindlimbs and suppressed CGRP expressions in DRG neurons of osteoporotic models. The novelty of this research suggests that IL-6 is one of the causes of immobility-induced osteoporotic pain regardless improvement of bone loss.
Collapse
|
29
|
Li Y, Lu L, Xie Y, Chen X, Tian L, Liang Y, Li H, Zhang J, Liu Y, Yu X. Interleukin-6 Knockout Inhibits Senescence of Bone Mesenchymal Stem Cells in High-Fat Diet-Induced Bone Loss. Front Endocrinol (Lausanne) 2020; 11:622950. [PMID: 33679606 PMCID: PMC7933660 DOI: 10.3389/fendo.2020.622950] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/31/2020] [Indexed: 02/05/2023] Open
Abstract
Obesity, a chronic low-grade inflammatory state, not only promotes bone loss, but also accelerates cell senescence. However, little is known about the mechanisms that link obesity, bone loss, and cell senescence. Interleukin-6 (IL-6), a pivotal inflammatory mediator increased during obesity, is a candidate for promoting cell senescence and an important part of senescence-associated secretory phenotype (SASP). Here, wild type (WT) and (IL-6 KO) mice were fed with high-fat diet (HFD) for 12 weeks. The results showed IL-6 KO mice gain less weight on HFD than WT mice. HFD induced trabecular bone loss, enhanced expansion of bone marrow adipose tissue (BMAT), increased adipogenesis in bone marrow (BM), and reduced the bone formation in WT mice, but it failed to do so in IL-6 KO mice. Furthermore, IL-6 KO inhibited HFD-induced clone formation of bone marrow cells (BMCs), and expression of senescence markers (p53 and p21). IL-6 antibody inhibited the activation of STAT3 and the senescence of bone mesenchymal stem cells (BMSCs) from WT mice in vitro, while rescued IL-6 induced senescence of BMSCs from IL-6 KO mice through the STAT3/p53/p21 pathway. In summary, our data demonstrated that IL-6 KO may maintain the balance between osteogenesis and adipogenesis in BM, and restrain senescence of BMSCs in HFD-induced bone loss.
Collapse
Affiliation(s)
- Yujue Li
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyun Lu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Xie
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Chen
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Tian
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Liang
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, China
| | - Huifang Li
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhang
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xijie Yu, ;
| |
Collapse
|
30
|
Mechanisms Underlying Bone Loss Associated with Gut Inflammation. Int J Mol Sci 2019; 20:ijms20246323. [PMID: 31847438 PMCID: PMC6940820 DOI: 10.3390/ijms20246323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with gastrointestinal diseases frequently suffer from skeletal abnormality, characterized by reduced bone mineral density, increased fracture risk, and/or joint inflammation. This pathological process is characterized by altered immune cell activity and elevated inflammatory cytokines in the bone marrow microenvironment due to disrupted gut immune response. Gastrointestinal disease is recognized as an immune malfunction driven by multiple factors, including cytokines and signaling molecules. However, the mechanism by which intestinal inflammation magnified by gut-residing actors stimulates bone loss remains to be elucidated. In this article, we discuss the main risk factors potentially contributing to intestinal disease-associated bone loss, and summarize current animal models, illustrating gut-bone axis to bridge the gap between intestinal inflammation and skeletal disease.
Collapse
|
31
|
Taves S, Sun J, Livingston EW, Chen X, Amiaud J, Brion R, Hannah WB, Bateman TA, Heymann D, Monahan PE. Hemophilia A and B mice, but not VWF -/-mice, display bone defects in congenital development and remodeling after injury. Sci Rep 2019; 9:14428. [PMID: 31594977 PMCID: PMC6783554 DOI: 10.1038/s41598-019-50787-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 09/19/2019] [Indexed: 12/31/2022] Open
Abstract
While joint damage is the primary co-morbidity of hemophilia, osteoporosis and osteopenia are also observed. Coagulation factor VIII deficient (FVIII−/−) mice develop an osteoporotic phenotype in the absence of induced hemarthrosis that is exacerbated two weeks after an induced joint injury. Here we have compared comprehensively the bone health of clotting factor VIII, factor IX, and Von Willebrand Factor knockout (FVIII−/−, FIX−/−, and VWF−/− respectively) mice both in the absence of joint hemorrhage and following induced joint injury. We found FVIII−/− and FIX−/− mice, but not VWF−/− mice, developmentally have an osteoporotic phenotype. Unilateral induced hemarthrosis causes further bone damage in both FVIII−/− and FIX−/− mice, but has little effect on VWF−/− bone health, indicating that the FVIII.VWF complex is not required for normal bone remodeling in vivo. To further investigate the bone healing following hemarthrosis in hemophilia we examined a two week time course using microCT, serum chemistry, and histological analysis. Elevated ratio of osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL), increased osterix+ osteoblastic cells, and decreased smoothness of the cortical bone surface were evident within several days of injury, indicative of acute heterotopic mineralization along the cortical surface. This was closely followed by increased interleukin-6 (IL-6) levels, increased osteoclast numbers, and significant trabecular bone loss. Uncoupled and disorganized bone formation and resorption continued for the duration of the study resulting in significant deterioration of the joint. Further elucidation of the shared mechanisms underlying abnormal bone homeostasis in the absence of FVIII or FIX is needed to guide evidence-based approaches to the screening and treatment of the prevalent bone defects in hemophilia A and B.
Collapse
Affiliation(s)
- Sarah Taves
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.,Global Research, Novo Nordisk A/S, Maløv, Denmark
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA.,Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Eric W Livingston
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA
| | - Xin Chen
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jerome Amiaud
- INSERM, U1238, Faculty of Medicine, Université de Nantes, Nantes, F-44093, France
| | - Regis Brion
- INSERM, U1238, Faculty of Medicine, Université de Nantes, Nantes, F-44093, France
| | - William B Hannah
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ted A Bateman
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Dominique Heymann
- INSERM, U1232, CRCiNA, Institut de Cancérologie de l'Ouest, Université de Nantes, Université d'Angers, Saint-Herblain, F-44805, France. .,University of Sheffield, INSERM, Associated European Laboratory Sarcoma Research Unit, Department of Oncology and Metabolism, Sheffield, S10 2RX, UK.
| | - Paul E Monahan
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA. .,Harold R. Roberts Comprehensive Hemophilia Diagnosis and Treatment Center, University of North Carolina, Chapel Hill, NC, USA. .,Spark Therapeutics, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Liu X, D'Cruz AA, Hansen J, Croker BA, Lawlor KE, Sims NA, Wicks IP. Deleting Suppressor of Cytokine Signaling-3 in chondrocytes reduces bone growth by disrupting mitogen-activated protein kinase signaling. Osteoarthritis Cartilage 2019; 27:1557-1563. [PMID: 31176017 DOI: 10.1016/j.joca.2019.05.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 05/01/2019] [Accepted: 05/29/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the impact of deleting Suppressor of Cytokine Signaling (SOCS)-3 (SOCS3) in chondrocytes during murine skeletal development. METHOD Mice with a conditional Socs3 allele (Socs3fl/fl) were crossed with a transgenic mouse expressing Cre recombinase under the control of the type II collagen promoter (Col2a1) to generate Socs3Δ/Δcol2 mice. Skeletal growth was analyzed over the lifespan of Socs3Δ/Δcol2 mice and controls by detailed histomorphology. Bone size and cortical bone development was evaluated by micro-computed tomography (micro-CT). Growth plate (GP) zone width, chondrocyte proliferation and apoptosis were assessed by immunofluorescence staining for Ki67 and TUNEL. Fibroblast growth factor receptor-3 (FGFR3) signaling in the GP was assessed by immunohistochemistry, while the effect of SOCS3 overexpression on FGFR3-driven pMAPK signaling in HEK293T cells was evaluated by Western blot. RESULTS Socs3Δ/Δcol2 mice of both sexes were consistently smaller compared to littermate controls throughout life. This phenotype was due to reduced long bone size, poor cortical bone development, reduced Ki67+ proliferative chondrocytes and decreased proliferative zone (PZ) width in the GP. Expression of pMAPK, but not pSTAT3, was increased in the GPs of Socs3Δ/Δcol2 mice relative to littermate controls. Overexpression of FGFR3 in HEK293T cells increased Fibroblast Growth Factor 18 (FGF18)-dependent Mitogen-activated protein kinase (MAPK) phosphorylation, while concomitant expression of SOCS3 reduced FGFR3 expression and abrogated MAPK signaling. CONCLUSION Our results suggest a potential role for SOCS3 in GP chondrocyte proliferation by regulating FGFR3-dependent MAPK signaling in response to FGF18.
Collapse
Affiliation(s)
- X Liu
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia; University of Queensland, Diamantina Institute, Brisbane, Queensland, 4102, Australia
| | - A A D'Cruz
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - J Hansen
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - B A Croker
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - K E Lawlor
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia; Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, 3168, Australia
| | - N A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia; Department of Medicine at St Vincent's Hospital, The University of Melbourne, 3065, Australia
| | - I P Wicks
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia; Rheumatology Unit, Royal Melbourne Hospital, Parkville, Victoria, 3050, Australia.
| |
Collapse
|
33
|
Sims NA, Martin TJ. Osteoclasts Provide Coupling Signals to Osteoblast Lineage Cells Through Multiple Mechanisms. Annu Rev Physiol 2019; 82:507-529. [PMID: 31553686 DOI: 10.1146/annurev-physiol-021119-034425] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bone remodeling is essential for the repair and replacement of damaged and old bone. The major principle underlying this process is that osteoclast-mediated resorption of a quantum of bone is followed by osteoblast precursor recruitment; these cells differentiate to matrix-producing osteoblasts, which form new bone to replace what was resorbed. Evidence from osteopetrotic syndromes indicate that osteoclasts not only resorb bone, but also provide signals to promote bone formation. Osteoclasts act upon osteoblast lineage cells throughout their differentiation by facilitating growth factor release from resorbed matrix, producing secreted proteins and microvesicles, and expressing membrane-bound factors. These multiple mechanisms mediate the coupling of bone formation to resorption in remodeling. Additional interactions of osteoclasts with osteoblast lineage cells, including interactions with canopy and reversal cells, are required to achieve coordination between bone formation and resorption during bone remodeling.
Collapse
Affiliation(s)
- Natalie A Sims
- Bone Cell Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; , .,Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - T John Martin
- Bone Cell Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; , .,Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia
| |
Collapse
|
34
|
Real SAS, Parveen F, Rehman AU, Shaik R, Deo SVS, Husain SA. Mutation, methylation and expression analysis of LIFR gene in Indian breast cancer patients. Mutat Res 2019; 816-818:111677. [PMID: 31557600 DOI: 10.1016/j.mrfmmm.2019.111677] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/10/2019] [Accepted: 08/02/2019] [Indexed: 01/19/2023]
Abstract
LIFR functions as a tumor suppressor and metastatic suppressor of breast cancer. The present study investigates the status of LIFR gene in Indian breast cancer patients. A total of 137 breast cancer tissue and 137 adjacent normal tissue which served as controls were analyzed for mutation by automated DNA sequencing, methylation through methylation-specific polymerase chain reaction and its corresponding expression at mRNA and protein level using real-time quantitative polymerase chain reaction and immunohistochemistry respectively in Indian breast cancer patients. All the molecular findings were statistically correlated with clinopathological parameters of the patients to identify its association. LIFR mRNA expression was found to be 2.534 ± 3.52 fold downregulated with subsequent absence of protein in 67.15% cases (92/137). The absence of LIFR protein coincided with 80.95% (85/105) methylated cases thereby showing a very strong correlation among the LIFR promoter methylation and LIFR protein expression (p = 0.0001). We also observed G2968C nucleotide change in 6/137 cases of exon 20 of LIFR gene resulting in Glu990Gln mutation. Correlation of LIFR promoter methylation with geographic location and age at menopause and LIFR mutation with age at menarche, age at first live birth, molecular subtypes of breast cancer, and lymph node status remained significant even after bonferroni correction (p ≤ 0.0027). All these data suggests the relevance of these associations in relation to Indian breast cancer patients. The loss of LIFR protein was frequently found in Indian breast cancer patients, and aberrant promoter methylation showed a significant correlation with its downregulation.
Collapse
Affiliation(s)
| | - Farah Parveen
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Asad Ur Rehman
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | | | - S V S Deo
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | | |
Collapse
|
35
|
Lundmark A, Hu YOO, Huss M, Johannsen G, Andersson AF, Yucel-Lindberg T. Identification of Salivary Microbiota and Its Association With Host Inflammatory Mediators in Periodontitis. Front Cell Infect Microbiol 2019; 9:216. [PMID: 31281801 PMCID: PMC6598052 DOI: 10.3389/fcimb.2019.00216] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/05/2019] [Indexed: 12/22/2022] Open
Abstract
Periodontitis is a microbial-induced chronic inflammatory disease, which may not only result in tooth loss, but can also contribute to the development of various systemic diseases. The transition from healthy to diseased periodontium depends on microbial dysbiosis and impaired host immune response. Although periodontitis is a common disease as well as associated with various systemic inflammatory conditions, the taxonomic profiling of the salivary microbiota in periodontitis and its association with host immune and inflammatory mediators has not been reported. Therefore, the aim of this study was to identify key pathogens and their potential interaction with the host's inflammatory mediators in saliva samples for periodontitis risk assessment. The microbial 16S rRNA gene sequencing and the levels of inflammatory mediators were performed in saliva samples from patients with chronic periodontitis and periodontally healthy control subjects. The salivary microbial community composition differed significantly between patients with chronic periodontitis and healthy controls. Our analyses identified a number of microbes, including bacteria assigned to Eubacterium saphenum, Tannerella forsythia, Filifactor alocis, Streptococcus mitis/parasanguinis, Parvimonas micra, Prevotella sp., Phocaeicola sp., and Fretibacterium sp. as more abundant in periodontitis, compared to healthy controls. In samples from healthy individuals, we identified Campylobacter concisus, and Veillonella sp. as more abundant. Integrative analysis of the microbiota and inflammatory mediators/cytokines revealed associations that included positive correlations between the pathogens Treponema sp. and Selenomas sp. and the cytokines chitinase 3-like 1, sIL-6Rα, sTNF-R1, and gp130/sIL-6Rβ. In addition, a negative correlation was identified between IL-10 and Filifactor alocis. Our results reveal distinct and disease-specific patterns of salivary microbial composition between patients with periodontitis and healthy controls, as well as significant correlations between microbiota and host-mediated inflammatory cytokines. The positive correlations between the pathogens Treponema sp. and Selenomas sp. and the cytokines chitinase 3-like 1, sIL-6Rα, sTNF-R1, and gp130/sIL-6Rβ might have the future potential to serve as a combined bacteria-host salivary biomarker panel for diagnosis of the chronic infectious disease periodontitis. However, further studies are required to determine the capacity of these microbes and inflammatory mediators as a salivary biomarker panel for periodontitis.
Collapse
Affiliation(s)
- Anna Lundmark
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Yue O O Hu
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Karolinska Institutet, Stockholm, Sweden
| | - Mikael Huss
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Gunnar Johannsen
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Anders F Andersson
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tülay Yucel-Lindberg
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
36
|
Lerner UH, Kindstedt E, Lundberg P. The critical interplay between bone resorbing and bone forming cells. J Clin Periodontol 2019; 46 Suppl 21:33-51. [DOI: 10.1111/jcpe.13051] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/05/2018] [Accepted: 12/01/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Ulf H. Lerner
- Centre for Bone and Arthritis Research at Department of Internal Medicine and Clinical Nutrition; Institute of Medicine; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
- Department of Odontology; Division of Molecular Periodontology; Umeå University; Umeå Sweden
| | - Elin Kindstedt
- Department of Odontology; Division of Molecular Periodontology; Umeå University; Umeå Sweden
| | - Pernilla Lundberg
- Department of Odontology; Division of Molecular Periodontology; Umeå University; Umeå Sweden
| |
Collapse
|
37
|
Persson E, Souza PPC, Floriano-Marcelino T, Conaway HH, Henning P, Lerner UH. Activation of Shc1 Allows Oncostatin M to Induce RANKL and Osteoclast Formation More Effectively Than Leukemia Inhibitory Factor. Front Immunol 2019; 10:1164. [PMID: 31191537 PMCID: PMC6547810 DOI: 10.3389/fimmu.2019.01164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/08/2019] [Indexed: 11/16/2022] Open
Abstract
Background and Purpose: The gp130 family of cytokines signals through receptors dimerizing with the gp130 subunit. Downstream signaling typically activates STAT3 but also SHP2/Ras/MAPK pathways. Oncostatin M (OSM) is a unique cytokine in this family since the receptor (OSMR) activates a non-redundant signaling pathway by recruitment of the adapter Shc1. We have studied the functional relevance of Shc1 for OSM-induced bone resorption. Experimental Approach: Osteoblasts were stimulated with OSM and STAT3 and Shc1 activations were studied using real-time PCR and Western blots. The role of STAT3 and Shc1 for OSM-induced RANKL expression and osteoclast formation was studied by silencing their mRNA expressions. Effects of OSM were compared to those of the closely related cytokine leukemia inhibitory factor (LIF). Key Results: OSM, but not LIF, induced the mRNA and protein expression of Shc1 and activated phosphorylation of Shc1 in the osteoblasts. Silencing of Shc1 decreased OSM-induced activation of STAT3 and RANKL expression. Silencing of STAT3 had no effect on activation of Shc1, but prevented the OSM-mediated increase of RANKL expression. Silencing of either Shc1 or STAT3 in osteoblasts decreased formation of osteoclasts in OSM-stimulated co-cultures of osteoblasts and macrophages. In agreement with these observations, OSM was a more potent and robust stimulator than LIF of RANKL formation and bone resorption in mouse calvariae and osteoclast formation in bone marrow cultures. Conclusions and Implications: Activation of the Shc1-dependent STAT3 signaling is crucial for OSM-induced osteoclast formation. Inhibition of Shc1 is a potential mechanism to specifically inhibit OSM-induced bone resorption.
Collapse
Affiliation(s)
- Emma Persson
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Pedro P C Souza
- Bone Biology Research Group, Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil.,School of Dentistry, Federal University of Goiás, Goiânia, Brazil
| | - Thais Floriano-Marcelino
- Bone Biology Research Group, Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Howard Herschel Conaway
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden.,Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
38
|
Zhong C, Szollosi D, Sun J, Hua B, Ghoneim O, Bill A, Zhuang Y, Edafiogho I. Novel Piperazino-Enaminones Decrease Pro-inflammatory Cytokines Following Hemarthrosis in a Hemophilia Mouse Model. Inflammation 2019; 42:1719-1729. [DOI: 10.1007/s10753-019-01032-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
39
|
Kim BJ, Koh JM. Coupling factors involved in preserving bone balance. Cell Mol Life Sci 2019; 76:1243-1253. [PMID: 30515522 PMCID: PMC11105749 DOI: 10.1007/s00018-018-2981-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/14/2018] [Accepted: 11/26/2018] [Indexed: 12/11/2022]
Abstract
Coupling during bone remodeling refers to the spatial and temporal coordination of bone resorption with bone formation. Studies have assessed the subtle interactions between osteoclasts and osteoblasts to preserve bone balance. Traditionally, coupling research related to osteoclast function has focused on bone resorption activity causing the release of growth factors embedded in the bone matrix. However, considerable evidence from in vitro, animal, and human studies indicates the importance of the osteoclasts themselves in coupling phenomena, and many osteoclast-derived coupling factors have been identified. These include sphingosine-1-phosphate, vesicular-receptor activator of nuclear factor-κB, collagen triple helix repeat containing 1, and cardiotrophin-1. Interestingly, neuronal guidance molecules, such as slit guidance ligand 3, semaphorin (SEMA) 3A, SEMA4D, and netrin-1, originally identified as instructive cues allowing the navigation of growing axons to their targets, have been shown to be involved in the intercellular cross-talk among bone cells. This review discusses osteoclast-osteoblast coupling signals, including recent advances and the potential roles of these signals as therapeutic targets for osteoporosis and as biomarkers predicting human bone health.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
40
|
Yuan B, Raucci MG, Fan Y, Zhu X, Yang X, Zhang X, Santin M, Ambrosio L. Injectable strontium-doped hydroxyapatite integrated with phosphoserine-tethered poly(epsilon-lysine) dendrons for osteoporotic bone defect repair. J Mater Chem B 2018; 6:7974-7984. [PMID: 32255042 DOI: 10.1039/c8tb02526f] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The control of the inflammatory response induced by the implantation of foreign biomaterials is fundamental in determining tissue healing. It has been shown that the activation of specific macrophage pathways upon contact with a biomaterial can lead either to a chronic inflammation preventing a physiological tissue repair or to an improved tissue healing. In the case of bone repair, calcium phosphate cements with good osteoconductivity properties have been successfully applied in bone defect filling and repair, but poor handling properties, insufficient viscous flow and unmatched degradation rate are still problems that remain unsolved. In this study, a strontium-doped hydroxyapatite (HA) gel was modified by integrating branched poly(epsilon-lysine) dendrons with third-generation branches exposing phosphoserine (SrHA/G3-K PS). The interaction of this material with macrophages was investigated in vitro, focusing on the secretion and gene expression of specific pro-inflammatory cytokines. Our results showed that the addition of strontium and G3-K PS to HA sol-gel could down-regulate the gene expression of inflammatory factors such as IL-1β, TNF-α and MCP-1, while increasing the gene expression of IL-6, a cytokine known for its osteogenic effect. These results were further confirmed by ELISA test of the respective protein concentrations. When exposed to supernatants of macrophage culture in the presence of strontium and G3-K PS, osteoblast viability was promoted with elevated osteogenic gene markers, in terms of OPG, ALP, OCN and COL-I. In vivo implantation experiments using an osteoporotic rat model with bone defect further confirmed that the addition of G3-K PS to HA could dramatically promote new bone regeneration. Although the introduction of strontium improved the degradation properties of the injectable materials, no positive effect on promoting in vivo bone regeneration was observed.
Collapse
Affiliation(s)
- Bo Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Chhana A, Pool B, Callon KE, Tay ML, Musson D, Naot D, McCarthy G, McGlashan S, Cornish J, Dalbeth N. Monosodium urate crystals reduce osteocyte viability and indirectly promote a shift in osteocyte function towards a proinflammatory and proresorptive state. Arthritis Res Ther 2018; 20:208. [PMID: 30201038 PMCID: PMC6131786 DOI: 10.1186/s13075-018-1704-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background Bone erosion is a frequent complication of gout and is strongly associated with tophi, which are lesions comprising inflammatory cells surrounding collections of monosodium urate (MSU) crystals. Osteocytes are important cellular mediators of bone remodeling. The aim of this study was to investigate the direct effects of MSU crystals and indirect effects of MSU crystal-induced inflammation on osteocytes. Methods For direct assays, MSU crystals were added to MLO-Y4 osteocyte cell line cultures or primary mouse osteocyte cultures. For indirect assays, the RAW264.7 macrophage cell line was cultured with or without MSU crystals, and conditioned medium from these cultures was added to MLO-Y4 cells. MLO-Y4 cell viability was assessed using alamarBlue® and LIVE/DEAD® assays, and MLO-Y4 cell gene expression and protein expression were assessed by real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA), respectively. Histological analysis was used to examine the relationship between MSU crystals, inflammatory cells, and osteocytes in human joints affected by tophaceous gout. Results In direct assays, MSU crystals reduced MLO-Y4 cell and primary mouse osteocyte viability but did not alter MLO-Y4 cell gene expression. In contrast, conditioned medium from MSU crystal-stimulated RAW264.7 macrophages did not affect MLO-Y4 cell viability but significantly increased MLO-Y4 cell expression of osteocyte-related factors including E11, connexin 43, and RANKL, and inflammatory mediators such as interleukin (IL)-6, IL-11, tumor necrosis factor (TNF)-α and cyclooxygenase-2 (COX-2). Inhibition of COX-2 in MLO-Y4 cells significantly reduced the indirect effects of MSU crystals. In histological analysis, CD68+ macrophages and MSU crystals were identified in close proximity to osteocytes within bone. COX-2 expression was also observed in tophaceous joint samples. Conclusions MSU crystals directly inhibit osteocyte viability and, through interactions with macrophages, indirectly promote a shift in osteocyte function that favors bone resorption and inflammation. These interactions may contribute to disordered bone remodeling in gout. Electronic supplementary material The online version of this article (10.1186/s13075-018-1704-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashika Chhana
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - Bregina Pool
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - Karen E Callon
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - Mei Lin Tay
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - David Musson
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - Dorit Naot
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - Geraldine McCarthy
- Department of Rheumatology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Susan McGlashan
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Jillian Cornish
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Bone & Joint Research Group, University of Auckland, Auckland, New Zealand. .,Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, New Zealand.
| |
Collapse
|
42
|
Lazzaro L, Tonkin BA, Poulton IJ, McGregor NE, Ferlin W, Sims NA. IL-6 trans-signalling mediates trabecular, but not cortical, bone loss after ovariectomy. Bone 2018; 112:120-127. [PMID: 29679733 DOI: 10.1016/j.bone.2018.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/04/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022]
Abstract
Bone loss associated with estrogen deficiency occurs due to a high level of bone remodelling, with a greater increase in the level of osteoclast-mediated bone resorption than osteoblast-mediated bone formation. Early studies showed that Interleukin-6 (IL-6) inhibition could prevent the increase in osteoclast numbers associated with ovariectomy. However, IL-6 signals through two possible pathways: classic IL-6 signalling (cis) utilizes a membrane-bound IL-6 receptor (IL-6R), while IL-6 trans-signalling occurs through a soluble IL-6R (sIL-6R). It is not known which of these pathways mediates the bone loss after ovariectomy. We therefore sought to determine whether specific pharmacological inhibition of IL-6 trans-signalling could prevent ovariectomy-induced bone loss in mice. We report that IL-6 trans-signalling inhibition prevented the increase in osteoclasts, and trabecular bone loss, associated with ovariectomy. IL-6 trans-signalling inhibition also reduced bone formation rate, but did not prevent the increase in osteoblast numbers. In contrast, cortical bone loss was not prevented by any IL-6 signalling inhibitor. This suggests that local production of sIL-6R mediates trabecular bone loss in estrogen deficiency, but the increased cortical bone resorption that leads to marrow expansion is independent of IL-6 signalling.
Collapse
Affiliation(s)
- Leah Lazzaro
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Brett A Tonkin
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Ingrid J Poulton
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Narelle E McGregor
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | | | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia.
| |
Collapse
|
43
|
Brook N, Brook E, Dharmarajan A, Dass CR, Chan A. Breast cancer bone metastases: pathogenesis and therapeutic targets. Int J Biochem Cell Biol 2018; 96:63-78. [DOI: 10.1016/j.biocel.2018.01.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/31/2017] [Accepted: 01/04/2018] [Indexed: 01/03/2023]
|
44
|
Liu Y, Song F, Wu S, He S, Meng M, Lv C, Yang Q, Chen S. Protein and mRNA expressions of IL-6 and its key signaling factors under orthodontic forces in mice: An in-vivo study. Am J Orthod Dentofacial Orthop 2017; 152:654-662. [PMID: 29103443 DOI: 10.1016/j.ajodo.2017.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 03/01/2017] [Accepted: 03/01/2017] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The purpose of this study was to investigate the mechanical loading-induced changes in protein and mRNA expressions of interleukin-6 (IL-6) and its key signaling factors glycoprotein 130 (gp130), signal transducer and activator of transcription 3 (STAT3), and the Src homology phosphotyrosine phosphatase (SHP2) at the tension and compression sides of the teeth in mouse models. METHODS A total of 55 C57B/6 mice (10 weeks old) were divided into 3 groups. Orthodontic force was applied in group A (experimental group, n = 30); the tooth movement device was placed without activation in group B (sham control group, n = 15), and group C (blank control group, n = 10). Tooth movement was induced by a nickel-titanium coil spring inserted between the maxillary left incisor and the first molar with a force of approximately 4 g. The animals were killed 12 days after the interventions; protein and mRNA expressions of IL-6, gp130, STAT3, and SHP2 in the periodontal tissues were observed with immunohistochemistry and in-situ hybridization, respectively. RESULTS In contrast with the control groups, we observed enhanced expressions of IL-6, gp130, STAT3, and SHP2 protein and mRNA at the mesial and distal sides of the teeth with application of orthodontic forces in the experimental group. In contrast with the distal side, we observed enhanced expression of gp130 protein and mRNA at the mesial side in the experimental group. CONCLUSIONS We observed enhanced expression of IL-6 and its key signaling factors gp130, STAT3, and SHP2 protein and mRNA at the tension and compression sides of the teeth with application of orthodontic forces. The mechanical loading applied for orthodontic tooth movement might induce changes in protein localization and mRNA expression patterns of IL-6 and its key signaling factors gp130, STAT3, and SHP2 at the tension and compression sides of the periodontal ligaments of the teeth in mouse models. The result might demonstrate the special role of IL-6 and its key signaling factors in the alveolar bone-modeling process.
Collapse
Affiliation(s)
- Yi Liu
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Fang Song
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shu Wu
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shushu He
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Mingmei Meng
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunxiao Lv
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qingqing Yang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
45
|
Bitra A, Rani BJ, Agarkar SS, Parihar AS, Vynath GP, Grover S. Gingival Crevicular Fluid Turnover Markers in Premenopausal vs Postmenopausal Women receiving Orthodontic Treatment. J Contemp Dent Pract 2017; 18:933-936. [PMID: 28989133 DOI: 10.5005/jp-journals-10024-2152] [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] [Indexed: 11/23/2022]
Abstract
BACKGROUND Orthodontic treatment is one of the commonly used dental treatments. Orthodontic forces act on the bone by modulating the biomolecules, chiefly the osteoprotegerin (OPG), osteopontin (OPN), receptor activator of nuclear factor kappa-B (RANK), and RANK ligand (RANKL) (OPG ligand). Hormonal changes are known to cause marked alteration in the levels of these biomolecules. Hence, we planned this study to evaluate the response of bone biomarkers in the gingival crevicular fluid (GCF) in postmenopausal women undergoing fixed orthodontic therapy. MATERIALS AND METHODS This study included assessment of 50 subjects who underwent orthodontic treatment from June 2012 to July 2016. All the patients were divided into two study groups with 25 patients in each group: premenopausal group and postmenopausal group. Similar orthodontic wires were used for controlling the forces applied in subjects of both the study groups and their GCF levels of RANKL, and OPN was assessed at baseline and 24 hours after the activation of orthodontic forces. All the results were compiled, assessed, and analyzed by Statistical Package for the Social Sciences software version 16.0. Chi-square test, Student's t-test, and Mann-Whitney U test were used for the assessment of the level of significance. RESULTS The mean values of RANKL and OPN in the premenopausal and postmenopausal groups were found to be 241.52 and 317.15 pg/μL respectively. The mean values of RANKL at baseline in the premenopausal and postmenopausal groups were found to be 7.15 and 3.84 pg/μL respectively. Nonsignificant results were obtained while comparing mean OPN and RANKL level alteration in between the two study groups. CONCLUSION The mean alterations in the GCF levels of bone biomarkers are similar for both premenopausal and postmeno-pausal women. CLINICAL SIGNIFICANCE For women with either premenopausal or postmenopausal status, orthodontic treatment appears to be equally safer.
Collapse
Affiliation(s)
- Anusha Bitra
- Department of Orthodontics and Dentofacial Orthopaedics Drs Sudha and Nageswara Rao Siddhartha Institute of Dental Sciences, Vijayawada, Andhra Pradesh, India
| | - B Jhansi Rani
- Department of Orthodontics and Dentofacial Orthopaedics Drs Sudha and Nageswara Rao Siddhartha Institute of Dental Sciences, Vijayawada, Andhra Pradesh, India
| | - Sanket S Agarkar
- Department of Orthodontics, Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj S Parihar
- Department of Periodontology, RKDF Dental College and Research Centre, Bhopal, Madhya Pradesh, India
| | - Gopinath P Vynath
- Department of Periodontics, Malabar Dental College & Research Center, Edappal, Kerala, India
| | - Shekhar Grover
- Department of Public Health Dentistry, Maulana Azad Dental College & Hospital, New Delhi, India, Phone: +919501544877, e-mail:
| |
Collapse
|
46
|
Smuthkochorn S, Palomo JM, Hans MG, Jones CS, Palomo L. Gingival crevicular fluid bone turnover biomarkers: How postmenopausal women respond to orthodontic activation. Am J Orthod Dentofacial Orthop 2017; 152:33-37. [PMID: 28651765 DOI: 10.1016/j.ajodo.2016.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Bone turnover associated with orthodontic tooth movement is evidenced by increased bone turnover markers in gingival crevicular fluid (GCF). Postmenopausal women have an increased concentration of serum bone turnover markers. The filtrate of this serum makes up GCF, but little is known of the bone turnover around teeth in this cohort. The objective of this investigation was to compare the GCF bone turnover markers in premenopausal vs postmenopausal women receiving orthodontic treatment at baseline and at orthodontic activation. METHODS Twenty-eight women were enrolled in the study and separated into 2 groups: premenopausal (16) and postmenopausal (12). Bone turnover was evaluated by GCF at baseline and 24 hours after orthodontic appliance activation. GCF concentrations of RANKL and OPN were measured using ELISA. Baseline and change in concentrations were compared between groups. RESULTS Baseline RANKL and OPN were significantly different between the premenopausal and postmenopausal groups (P <0.05). Both markers increased significantly from baseline to 24 hours after orthodontic appliance activation in both groups (P <0.05). However, the response to orthodontic activation was not significantly different between groups. CONCLUSIONS Although postmenopausal women have a different bone turnover profile at baseline than do their premenopausal counterparts, there is no difference in their response to orthodontic activation. This confers a level of security associated with orthodontic activation. Future studies are warranted to construct biomarker curves throughout orthodontic therapy.
Collapse
Affiliation(s)
| | - J Martin Palomo
- Department of Orthodontics, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Mark G Hans
- Department of Orthodontics, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Leena Palomo
- Department of Periodontology, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
47
|
Cytokines in Endocrine Dysfunction of Plasma Cell Disorders. Mediators Inflamm 2017; 2017:7586174. [PMID: 28740334 PMCID: PMC5504949 DOI: 10.1155/2017/7586174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/25/2017] [Indexed: 12/25/2022] Open
Abstract
Monoclonal gammopathies (MG) are classically associated with lytic bone lesions, hypercalcemia, anemia, and renal insufficiency. However, in some cases, symptoms of endocrine dysfunction are more prominent than these classical signs and misdiagnosis can thus be possible. This concerns especially the situation where the presence of M-protein is limited and the serum protein electrophoresis (sPEP) appears normal. To understand the origin of the endocrine symptoms associated with MG, we overview here the current knowledge on the complexity of interactions between cytokines and the endocrine system in MG and discuss the perspectives for both the diagnosis and treatments for this class of diseases. We also illustrate the role of major cytokines and growth factors such as IL-6, IL-1β, TNF-α, and VEGF in the endocrine system, as these tumor-relevant signaling molecules not only help the clonal expansion and invasion of the tumor cells but also influence cellular metabolism through autocrine, paracrine, and endocrine mechanisms. We further discuss the broader impact of these tumor environment-derived molecules and proinflammatory state on systemic hormone signaling. The diagnostic challenges and clinical work-up are illustrated from the point of view of an endocrinologist.
Collapse
|
48
|
Kohart NA, Elshafae SM, Breitbach JT, Rosol TJ. Animal Models of Cancer-Associated Hypercalcemia. Vet Sci 2017; 4:vetsci4020021. [PMID: 29056680 PMCID: PMC5606604 DOI: 10.3390/vetsci4020021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 03/14/2017] [Accepted: 04/10/2017] [Indexed: 02/05/2023] Open
Abstract
Cancer-associated hypercalcemia (CAH) is a frequently-occurring paraneoplastic syndrome that contributes to substantial patient morbidity and occurs in both humans and animals. Patients with CAH are often characterized by markedly elevated serum calcium concentrations that result in a range of clinical symptoms involving the nervous, gastrointestinal and urinary systems. CAH is caused by two principle mechanisms; humorally-mediated and/or through local osteolytic bone metastasis resulting in excessive calcium release from resorbed bone. Humoral hypercalcemia of malignancy (HHM) is the most common mechanism and is due to the production and release of tumor-associated cytokines and humoral factors, such as parathyroid hormone-related protein (PTHrP), that act at distant sites to increase serum calcium concentrations. Local osteolytic hypercalcemia (LOH) occurs when primary or metastatic bone tumors act locally by releasing factors that stimulate osteoclast activity and bone resorption. LOH is a less frequent cause of CAH and in some cases can induce hypercalcemia in concert with HHM. Rarely, ectopic production of parathyroid hormone has been described. PTHrP-mediated hypercalcemia is the most common mechanism of CAH in human and canine malignancies and is recognized in other domestic species. Spontaneous and experimentally-induced animal models have been developed to study the mechanisms of CAH. These models have been essential for the evaluation of novel approaches and adjuvant therapies to manage CAH. This review will highlight the comparative aspects of CAH in humans and animals with a discussion of the available animal models used to study the pathogenesis of this important clinical syndrome.
Collapse
Affiliation(s)
- Nicole A Kohart
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Said M Elshafae
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Banha 13511, Egypt.
| | - Justin T Breitbach
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Thomas J Rosol
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
49
|
Hall MD, Murray CA, Valdez MJ, Perantoni AO. Mesoderm-specific Stat3 deletion affects expression of Sox9 yielding Sox9-dependent phenotypes. PLoS Genet 2017; 13:e1006610. [PMID: 28166224 PMCID: PMC5319801 DOI: 10.1371/journal.pgen.1006610] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 02/21/2017] [Accepted: 01/30/2017] [Indexed: 01/14/2023] Open
Abstract
To date, mutations within the coding region and translocations around the SOX9 gene both constitute the majority of genetic lesions underpinning human campomelic dysplasia (CD). While pathological coding-region mutations typically result in a non-functional SOX9 protein, little is known about what mechanism(s) controls normal SOX9 expression, and subsequently, which signaling pathways may be interrupted by alterations occurring around the SOX9 gene. Here, we report the identification of Stat3 as a key modulator of Sox9 expression in nascent cartilage and developing chondrocytes. Stat3 expression is predominant in tissues of mesodermal origin, and its conditional ablation using mesoderm-specific TCre, in vivo, causes dwarfism and skeletal defects characteristic of CD. Specifically, Stat3 loss results in the expansion of growth plate hypertrophic chondrocytes and deregulation of normal endochondral ossification in all bones examined. Conditional deletion of Stat3 with a Sox9Cre driver produces palate and tracheal irregularities similar to those described in Sox9+/- mice. Furthermore, mesodermal deletion of Stat3 causes global embryonic down regulation of Sox9 expression and function in vivo. Mechanistic experiments ex vivo suggest Stat3 can directly activate the expression of Sox9 by binding to its proximal promoter following activation. These findings illuminate a novel role for Stat3 in chondrocytes during skeletal development through modulation of a critical factor, Sox9. Importantly, they further provide the first evidence for the modulation of a gene product other than Sox9 itself which is capable of modeling pathological aspects of CD and underscore a potentially valuable therapeutic target for patients with the disorder.
Collapse
Affiliation(s)
- Michael D. Hall
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| | - Caroline A. Murray
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| | - Michael J. Valdez
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| | - Alan O. Perantoni
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| |
Collapse
|
50
|
Hao Z, Ma Y, Wu J, Li X, Chen H, Shen J, Wang H. Osteocytes regulate osteoblast differentiation and osteoclast activity through Interleukin-6 under mechanical loading. RSC Adv 2017. [DOI: 10.1039/c7ra09308j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Osteocytes are the major mechanosensors that respond to mechanical strain and regulate bone formation and resorption.
Collapse
Affiliation(s)
- Zhichao Hao
- Guanghua School of Stomatology
- Hospital of Stomatology
- Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Stomatology
- Guangzhou 510055
| | - Yuanyuan Ma
- Guanghua School of Stomatology
- Hospital of Stomatology
- Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Stomatology
- Guangzhou 510055
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province
- School of Engineering
- Sun Yat-sen University
- Guangzhou
- China
| | - Xianxian Li
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital
- Chengdu 610041
- China
| | - Helin Chen
- State Key Laboratory of Oral Diseases
- National Clinical Research Center for Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
| | - Jiefei Shen
- State Key Laboratory of Oral Diseases
- National Clinical Research Center for Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
| | - Hang Wang
- State Key Laboratory of Oral Diseases
- National Clinical Research Center for Oral Diseases
- West China Hospital of Stomatology
- Sichuan University
- Chengdu 610041
| |
Collapse
|