1
|
Babendreyer A, Kieselhorst J, Rinkens C, Lyashenko AM, Düsterhöft S, Jahr H, Craveiro RB, Wolf M, Ludwig A. Downregulation of the metalloproteinases ADAM10 or ADAM17 promotes osteoclast differentiation. Cell Commun Signal 2024; 22:322. [PMID: 38863060 PMCID: PMC11167776 DOI: 10.1186/s12964-024-01690-y] [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: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Bone resorption is driven through osteoclast differentiation by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-Β ligand (RANKL). We noted that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 are downregulated at the expression level during osteoclast differentiation of the murine monocytic cell line RAW264.7 in response to RANKL. Both proteinases are well known to shed a variety of single-pass transmembrane molecules from the cell surface. We further showed that inhibitors of ADAM10 or ADAM17 promote osteoclastic differentiation and furthermore enhance the surface expression of receptors for RANKL and M-CSF on RAW264.7 cells. Using murine bone marrow-derived monocytic cells (BMDMCs), we demonstrated that a genetic deficiency of ADAM17 or its required regulator iRhom2 leads to increased osteoclast development in response to M-CSF and RANKL stimulation. Moreover, ADAM17-deficient osteoclast precursor cells express increased levels of the receptors for RANKL and M-CSF. Thus, ADAM17 negatively regulates osteoclast differentiation, most likely through shedding of these receptors. To assess the time-dependent contribution of ADAM10, we blocked this proteinase by adding a specific inhibitor on day 0 of BMDMC stimulation with M-CSF or on day 7 of subsequent stimulation with RANKL. Only ADAM10 inhibition beginning on day 7 increased the size of developing osteoclasts indicating that ADAM10 suppresses osteoclast differentiation at a later stage. Finally, we could confirm our findings in human peripheral blood mononuclear cells (PBMCs). Thus, downregulation of either ADAM10 or ADAM17 during osteoclast differentiation may represent a novel regulatory mechanism to enhance their differentiation process. Enhanced bone resorption is a critical issue in osteoporosis and is driven through osteoclast differentiation by specific osteogenic mediators. The present study demonstrated that the metalloproteinases ADAM17 and ADAM10 critically suppress osteoclast development. This was observed for a murine cell line, for isolated murine bone marrow cells and for human blood cells by either preferential inhibition of the proteinases or by gene knockout. As a possible mechanism, we studied the surface expression of critical receptors for osteogenic mediators on developing osteoclasts. Our findings revealed that the suppressive effects of ADAM17 and ADAM10 on osteoclastogenesis can be explained in part by the proteolytic cleavage of surface receptors by ADAM10 and ADAM17, which reduces the sensitivity of these cells to osteogenic mediators. We also observed that osteoclast differentiation was associated with the downregulation of ADAM10 and ADAM17, which reduced their suppressive effects. We therefore propose that this downregulation serves as a feedback loop for enhancing osteoclast development.
Collapse
Affiliation(s)
- Aaron Babendreyer
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Julia Kieselhorst
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Cindy Rinkens
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Anastasia M Lyashenko
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Holger Jahr
- Institute of Anatomy and Cell Biology, University Hospital RWTH Aachen, Aachen, Germany
- Institute of Structural Mechanics and Lightweight Design, RWTH Aachen University, Aachen, Germany
| | - Rogerio B Craveiro
- Department of Orthodontics, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, University Hospital RWTH Aachen, Aachen, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
2
|
Rabinowitsch AI, Maretzky T, Weskamp G, Haxaire C, Tueshaus J, Lichtenthaler SF, Monette S, Blobel CP. Analysis of the function of ADAM17 in iRhom2 curly-bare and tylosis with esophageal cancer mutant mice. J Cell Sci 2023; 136:jcs260910. [PMID: 37282854 PMCID: PMC10357010 DOI: 10.1242/jcs.260910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/31/2023] [Indexed: 06/08/2023] Open
Abstract
Tylosis with oesophageal cancer (TOC) is a rare familial disorder caused by cytoplasmic mutations in inactive rhomboid 2 (iRhom2 or iR2, encoded by Rhbdf2). iR2 and the related iRhom1 (or iR1, encoded by Rhbdf1) are key regulators of the membrane-anchored metalloprotease ADAM17, which is required for activating EGFR ligands and for releasing pro-inflammatory cytokines such as TNFα (or TNF). A cytoplasmic deletion in iR2, including the TOC site, leads to curly coat or bare skin (cub) in mice, whereas a knock-in TOC mutation (toc) causes less severe alopecia and wavy fur. The abnormal skin and hair phenotypes of iR2cub/cub and iR2toc/toc mice depend on amphiregulin (Areg) and Adam17, as loss of one allele of either gene rescues the fur phenotypes. Remarkably, we found that iR1-/- iR2cub/cub mice survived, despite a lack of mature ADAM17, whereas iR2cub/cub Adam17-/- mice died perinatally, suggesting that the iR2cub gain-of-function mutation requires the presence of ADAM17, but not its catalytic activity. The iR2toc mutation did not substantially reduce the levels of mature ADAM17, but instead affected its function in a substrate-selective manner. Our findings provide new insights into the role of the cytoplasmic domain of iR2 in vivo, with implications for the treatment of TOC patients.
Collapse
Affiliation(s)
- Ariana I. Rabinowitsch
- Tri-Institutional MD/PhD Program, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, The Rockefeller University, New York, NY 10021, USA
- Program in Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Thorsten Maretzky
- Inflammation Program and Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Gisela Weskamp
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Coline Haxaire
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Johanna Tueshaus
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Institute for Advanced Study, Technical University of Munich, 85748 Garching, Germany
| | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Institute for Advanced Study, Technical University of Munich, 85748 Garching, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sébastien Monette
- Tri-Institutional Laboratory of Comparative Pathology, Hospital for Special Surgery, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, NY 10021, USA
| | - Carl P. Blobel
- Tri-Institutional MD/PhD Program, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, The Rockefeller University, New York, NY 10021, USA
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
- Institute for Advanced Study, Technical University of Munich, 85748 Garching, Germany
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
3
|
Mangiavini L, Peretti GM, Canciani B, Maffulli N. Epidermal growth factor signalling pathway in endochondral ossification: an evidence-based narrative review. Ann Med 2022; 54:37-50. [PMID: 34955078 PMCID: PMC8725985 DOI: 10.1080/07853890.2021.2015798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During endochondral bone development, a complex process that leads to the formation of the majority of skeletal elements, mesenchymal cells condense, differentiating into chondrocytes and producing the foetal growth plate. Chondrocytes progressively hypertrophy, induce angiogenesis and are then gradually replaced by bone. Epidermal Growth Factor (EGF), one of many growth factors, is the prototype of the EGF-ligand family, which comprises several proteins involved in cell proliferation, migration and survival. In bone, EGF pathway signalling finely tunes the first steps of chondrogenesis by maintaining mesenchymal cells in an undifferentiated stage, and by promoting hypertrophic cartilage replacement. Moreover, EGF signalling modulates bone homeostasis by stimulating osteoblast and osteoclast proliferation, and by regulating osteoblast differentiation under specific spatial and temporal conditions. This evidence-based narrative review describes the EGF pathway in bone metabolism and endochondral bone development. This comprehensive description may be useful in light of possible clinical applications in orthopaedic practice. A deeper knowledge of the role of EGF in bone may be useful in musculoskeletal conditions which may benefit from the modulation of this signalling pathway.Key messagesThe EGF pathway is involved in bone metabolism.EGF signalling is essential in the very early stages of limb development by maintaining cells in an undifferentiated stage.EGF pathway positively regulates chondrocyte proliferation, negatively modulates hypertrophy, and favours cartilage replacement by bone.EGF and EGF-like proteins finely tune the proliferation and differentiation of bone tissue cells, and they also regulate the initial phases of endochondral ossification.
Collapse
Affiliation(s)
- L Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - G M Peretti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical Sciences for Health, Università Degli Studi di Milano, Milan, Italy
| | - B Canciani
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - N Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy.,Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK.,School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent, UK
| |
Collapse
|
4
|
Zhao Y, Dávila EM, Li X, Tang B, Rabinowitsch AI, Perez-Aguilar JM, Blobel CP. Identification of Molecular Determinants in iRhoms1 and 2 That Contribute to the Substrate Selectivity of Stimulated ADAM17. Int J Mol Sci 2022; 23:12796. [PMID: 36361585 PMCID: PMC9654401 DOI: 10.3390/ijms232112796] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 09/29/2023] Open
Abstract
The metalloprotease ADAM17 is a key regulator of the TNFα, IL-6R and EGFR signaling pathways. The maturation and function of ADAM17 is controlled by the seven-membrane-spanning proteins iRhoms1 and 2. The functional properties of the ADAM17/iRhom1 and ADAM17/iRhom2 complexes differ, in that stimulated shedding of most ADAM17 substrates tested to date can be supported by iRhom2, whereas iRhom1 can only support stimulated shedding of very few ADAM17 substrates, such as TGFα. The first transmembrane domain (TMD1) of iRhom2 and the sole TMD of ADAM17 are important for the stimulated shedding of ADAM17 substrates by iRhom2. However, little is currently known about how the iRhoms interact with different substrates to control their stimulated shedding by ADAM17. To provide new insights into this topic, we tested how various chimeras between iRhom1 and iRhom2 affect the stimulated processing of the EGFR-ligands TGFα (iRhom1- or 2-dependent) and EREG (iRhom2-selective) by ADAM17. This uncovered an important role for the TMD7 of the iRhoms in determining their substrate selectivity. Computational methods utilized to characterize the iRhom1/2/substrate interactions suggest that the substrate selectivity is determined, at least in part, by a distinct accessibility of the substrate cleavage site to stimulated ADAM17. These studies not only provide new insights into why the substrate selectivity of stimulated iRhom2/ADAM17 differs from that of iRhom1/ADAM17, but also suggest new approaches for targeting the release of specific ADAM17 substrates.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Eliud Morales Dávila
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Xue Li
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Beiyu Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ariana I. Rabinowitsch
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Jose Manuel Perez-Aguilar
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Carl P. Blobel
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10021, USA
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| |
Collapse
|
5
|
Kaneko T, Horiuchi K, Chijimatsu R, Mori D, Nagata K, Omata Y, Yano F, Inui H, Moro T, Tanaka S, Saito T. Regulation of osteoarthritis development by ADAM17/Tace in articular cartilage. J Bone Miner Metab 2022; 40:196-207. [PMID: 34751824 DOI: 10.1007/s00774-021-01278-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION A disintegrin and metalloproteinase 17 (Adam17), also known as TNFα-converting enzyme (Tace), is a membrane-anchored protein involved in shedding of TNF, IL-6 receptor, ligands of epidermal growth factor receptor (EGFR), and Notch receptor. This study aimed to examine the role of Adam17 in adult articular cartilage and osteoarthritis (OA) pathophysiology. MATERIALS AND METHODS Adam17 expression was examined in mouse knee joints during OA development. We analyzed OA development in tamoxifen-inducible chondrocyte-specific Adam17 knockout mice of a resection of the medial meniscus and medial collateral ligament (medial) model, destabilization of the medial meniscus (DMM) model, and aging model. We analyzed downstream pathways by in vitro experiments, and further performed intra-articular administration of an Adam17 inhibitor TAPI-0 for surgically induced mouse OA. RESULTS Adam17 expression in mouse articular cartilage was increased by OA progression. In all models, Adam17 knockout mice showed ameliorated progression of articular cartilage degradation. Adam17 knockout decreased matrix metallopeptidase 13 (Mmp13) expression in both in vivo and in vitro experiments, whereas Adam17 activation by phorbol-12-myristate-13-acetate (PMA) increased Mmp13 and decreased aggrecan in mouse primary chondrocytes. Adam17 activation enhanced release of soluble TNF and transforming growth factor alpha, a representative EGF ligand, from mouse primary chondrocytes, while it did not change release of soluble IL-6 receptor or nuclear translocation of Notch1 intercellular domain. Intra-articular administration of the Adam17 inhibitor ameliorated OA progression. CONCLUSIONS This study demonstrates regulation of OA development by Adam17, involvement of EGFR and TNF pathways, and the possibility of Adam17 as a therapeutic target for OA.
Collapse
Affiliation(s)
- Taizo Kaneko
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, 359-8513, Japan
| | - Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Daisuke Mori
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kosei Nagata
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasunori Omata
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Fumiko Yano
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroshi Inui
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Toru Moro
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Division of Science for Joint Reconstruction, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
6
|
Lees-Shepard JB, Flint K, Fisher M, Omi M, Richard K, Antony M, Chen PJ, Yadav S, Threadgill D, Maihle NJ, Dealy CN. Cross-talk between EGFR and BMP signals regulates chondrocyte maturation during endochondral ossification. Dev Dyn 2021; 251:75-94. [PMID: 34773433 DOI: 10.1002/dvdy.438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Progressive maturation of growth plate chondrocytes drives long bone growth during endochondral ossification. Signals from the epidermal growth factor receptor (EGFR), and from bone morphogenetic protein-2 (BMP2), are required for normal chondrocyte maturation. Here, we investigated cross-talk between EGFR and BMP2 signals in developing and adult growth plates. RESULTS Using in vivo mouse models of conditional cartilage-targeted EGFR or BMP2 loss, we show that canonical BMP signal activation is increased in the hypertrophic chondrocytes of EGFR-deficient growth plates; whereas EGFR signal activation is increased in the reserve, prehypertrophic and hypertrophic chondrocytes of BMP2-deficient growth plates. EGFR-deficient chondrocytes displayed increased BMP signal activation in vitro, accompanied by increased expression of IHH, COL10A1, and RUNX2. Hypertrophic differentiation and BMP signal activation were suppressed in normal chondrocyte cultures treated with the EGFR ligand betacellulin, effects that were partially blocked by simultaneous treatment with BMP2 or a chemical EGFR antagonist. CONCLUSIONS Cross-talk between EGFR and BMP2 signals occurs during chondrocyte maturation. In the reserve and prehypertrophic zones, BMP2 signals unilaterally suppress EGFR activity; in the hypertrophic zone, EGFR and BMP2 signals repress each other. This cross-talk may play a role in regulating chondrocyte maturation in developing and adult growth plates.
Collapse
Affiliation(s)
- John B Lees-Shepard
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Kaitlyn Flint
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Melanie Fisher
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Minoru Omi
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Kelsey Richard
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Michelle Antony
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Po Jung Chen
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Sumit Yadav
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David Threadgill
- Department of Veterinary Pathology, Texas A&M University, College Station, Texas, USA.,Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, USA
| | - Nita J Maihle
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA.,Department of Cell & Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Caroline N Dealy
- Department of Orthodontics, University of Connecticut Health Center, Farmington, Connecticut, USA.,Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT, USA.,Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, USA.,Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
7
|
Lora J, Weskamp G, Li TM, Maretzky T, Shola DTN, Monette S, Lichtenthaler SF, Lu TT, Yang C, Blobel CP. Targeted truncation of the ADAM17 cytoplasmic domain in mice results in protein destabilization and a hypomorphic phenotype. J Biol Chem 2021; 296:100733. [PMID: 33957124 PMCID: PMC8191336 DOI: 10.1016/j.jbc.2021.100733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/28/2022] Open
Abstract
A disintegrin and metalloprotease 17 (ADAM17) is a cell-surface metalloprotease that serves as the principle sheddase for tumor necrosis factor α (TNFα), interleukin-6 receptor (IL-6R), and several ligands of the epidermal growth factor receptor (EGFR), regulating these crucial signaling pathways. ADAM17 activation requires its transmembrane domain, but not its cytoplasmic domain, and little is known about the role of this domain in vivo. To investigate, we used CRISPR-Cas9 to mutate the endogenous Adam17 locus in mice to produce a mutant ADAM17 lacking its cytoplasmic domain (Adam17Δcyto). Homozygous Adam17Δcyto animals were born at a Mendelian ratio and survived into adulthood with slightly wavy hair and curled whiskers, consistent with defects in ADAM17/EGFR signaling. At birth, Adam17Δcyto mice resembled Adam17−/− mice in that they had open eyes and enlarged semilunar heart valves, but they did not have bone growth plate defects. The deletion of the cytoplasmic domain resulted in strongly decreased ADAM17 protein levels in all tissues and cells examined, providing a likely cause for the hypomorphic phenotype. In functional assays, Adam17Δcyto mouse embryonic fibroblasts and bone-marrow-derived macrophages had strongly reduced ADAM17 activity, consistent with the reduced protein levels. Nevertheless, ADAM17Δcyto could be stimulated by PMA, a well-characterized posttranslational activator of ADAM17, corroborating that the cytoplasmic domain of endogenous ADAM17 is not required for its rapid response to PMA. Taken together, these results provide the first evidence that the cytoplasmic domain of ADAM17 plays a pivotal role in vivo in regulating ADAM17 levels and function.
Collapse
Affiliation(s)
- Jose Lora
- Physiology, Biophysics and Systems Biology Program, Weill Cornell Medicine, New York, New York, USA; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - Gisela Weskamp
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - Thomas M Li
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Thorsten Maretzky
- Inflammation Program and Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Dorjee T N Shola
- CRISPR and Genome Editing Resource Center, Rockefeller University, New York, New York, USA
| | - Sébastien Monette
- Tri-Institutional Laboratory of Comparative Pathology, Sloan-Kettering Institute, New York, New York, USA
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Technical University of Munich, Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Technical University of Munich, Munich, Germany; Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York, USA
| | - Chingwen Yang
- CRISPR and Genome Editing Resource Center, Rockefeller University, New York, New York, USA
| | - Carl P Blobel
- Physiology, Biophysics and Systems Biology Program, Weill Cornell Medicine, New York, New York, USA; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA; Institute for Advanced Study, Technical University of Munich, Garching, Germany; Department of Medicine, Weill Cornell Medicine, New York, New York, USA; Department of Biophysics, Physiology and Systems Biology, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
8
|
Fang R, Haxaire C, Otero M, Lessard S, Weskamp G, McIlwain DR, Mak TW, Lichtenthaler SF, Blobel CP. Role of iRhoms 1 and 2 in Endochondral Ossification. Int J Mol Sci 2020; 21:ijms21228732. [PMID: 33227998 PMCID: PMC7699240 DOI: 10.3390/ijms21228732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
Growth of the axial and appendicular skeleton depends on endochondral ossification, which is controlled by tightly regulated cell–cell interactions in the developing growth plates. Previous studies have uncovered an important role of a disintegrin and metalloprotease 17 (ADAM17) in the normal development of the mineralized zone of hypertrophic chondrocytes during endochondral ossification. ADAM17 regulates EGF-receptor signaling by cleaving EGFR-ligands such as TGFα from their membrane-anchored precursor. The activity of ADAM17 is controlled by two regulatory binding partners, the inactive Rhomboids 1 and 2 (iRhom1, 2), raising questions about their role in endochondral ossification. To address this question, we generated mice lacking iRhom2 (iR2−/−) with floxed alleles of iRhom1 that were specifically deleted in chondrocytes by Col2a1-Cre (iR1∆Ch). The resulting iR2−/−iR1∆Ch mice had retarded bone growth compared to iR2−/− mice, caused by a significantly expanded zone of hypertrophic mineralizing chondrocytes in the growth plate. Primary iR2−/−iR1∆Ch chondrocytes had strongly reduced shedding of TGFα and other ADAM17-dependent EGFR-ligands. The enlarged zone of mineralized hypertrophic chondrocytes in iR2−/−iR1∆Ch mice closely resembled the abnormal growth plate in A17∆Ch mice and was similar to growth plates in Tgfα−/− mice or mice with EGFR mutations. These data support a model in which iRhom1 and 2 regulate bone growth by controlling the ADAM17/TGFα/EGFR signaling axis during endochondral ossification.
Collapse
Affiliation(s)
- Renpeng Fang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China;
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021, USA; (C.H.); (G.W.)
| | - Coline Haxaire
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021, USA; (C.H.); (G.W.)
| | - Miguel Otero
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021, USA; (M.O.); (S.L.)
| | - Samantha Lessard
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021, USA; (M.O.); (S.L.)
| | - Gisela Weskamp
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021, USA; (C.H.); (G.W.)
| | - David R. McIlwain
- Baxter Laboratory in Stem Cell Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Tak W. Mak
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada;
| | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany;
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Institute for Advanced Study, Technische Universität München, 85748 Garching, Germany
| | - Carl P. Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery at Weill Cornell Medicine, New York, NY 10021, USA; (C.H.); (G.W.)
- Institute for Advanced Study, Technische Universität München, 85748 Garching, Germany
- Department of Medicine, Department of Biophysics, Physiology and Systems Biology, Weill Cornell Medicine, New York, NY 10021, USA
- Correspondence: ; Tel.: +212-606-1429; Fax: +212-774-2560
| |
Collapse
|
9
|
Status update on iRhom and ADAM17: It's still complicated. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1567-1583. [PMID: 31330158 DOI: 10.1016/j.bbamcr.2019.06.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
Several membrane-bound proteins with a single transmembrane domain are subjected to limited proteolysis at the cell surface. This cleavage leads to the release of their biologically active ectodomains, which can trigger different signalling pathways. In many cases, this ectodomain shedding is mediated by members of the family of a disintegrins and metalloproteinases (ADAMs). ADAM17 in particular is responsible for the cleavage of several proinflammatory mediators, growth factors, receptors and adhesion molecules. Due to its direct involvement in the release of these signalling molecules, ADAM17 can be positively and negatively involved in various physiological processes as well as in inflammatory, fibrotic and malignant pathologies. This central role of ADAM17 in a variety of processes requires strict multi-level regulation, including phosphorylation, various conformational changes and endogenous inhibitors. Recent research has shown that an early, crucial control mechanism is interaction with certain adapter proteins identified as iRhom1 and iRhom2, which are pseudoproteases of the rhomboid superfamily. Thus, iRhoms have also a decisive influence on physiological and pathophysiological signalling processes regulated by ADAM17. Their characteristic gene expression profiles, the specific consequences of gene knockouts and finally the occurrence of disease-associated mutations suggest that iRhom1 and iRhom2 undergo different gene regulation in order to fulfil their function in different cell types and are therefore only partially redundant. Therefore, there is not only interest in ADAM17, but also in iRhoms as therapeutic targets. However, to exploit the therapeutic potential, the regulation of ADAM17 activity and in particular its interaction with iRhoms must be well understood.
Collapse
|
10
|
Yang F, Lin ZW, Huang TY, Chen TT, Cui J, Li MY, Hua YQ. Ligustilide, a major bioactive component of Angelica sinensis, promotes bone formation via the GPR30/EGFR pathway. Sci Rep 2019; 9:6991. [PMID: 31061445 PMCID: PMC6502875 DOI: 10.1038/s41598-019-43518-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022] Open
Abstract
Angelica sinensis (Oliv.) Diels is a widely-used traditional Chinese herbal medicine in treating osteoporosis. Ligustilide (LIG) is the main component of A. sinensis and is considered to be the most effective biologically active ingredient in this plant. LIG has been found to have multiple pharmacological activities, such as anti-atherosclerosis, neuroprotection, anticancer, anti-inflammatory and analgesic. However, little is known regarding its anti-osteoporotic effects. The aims of this study were to investigate any protective effect of LIG on bone formation. The results showed that LIG significantly ameliorated inhibition of bone formation in zebrafish caused by prednisolone. LIG promoted osteoblast differentiation, including that of the pre-osteoblastic cell line MC3T3-E1 and bone marrow mesenchymal stem cells. LIG greatly improved the viability of MC3T3-E1 cells exposed to H2O2, attenuated H2O2-induced apoptosis and increased the expression of Bcl-2. Furthermore, LIG treatment lead to marked activation of phosphorylated EGFR and ERK1/2. These effects could be obviously inhibited by blocking GPR30 signaling with the specific inhibitor G15. Collectively, the results reveal that GPR30 is a positive switch for LIG to increase bone formation via regulation of EGFR, and these results provide evidence for the potential of LIG to treat osteoporosis.
Collapse
Affiliation(s)
- F Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Z W Lin
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - T Y Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - T T Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - J Cui
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - M Y Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Y Q Hua
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China.
| |
Collapse
|
11
|
Qin L, Beier F. EGFR Signaling: Friend or Foe for Cartilage? JBMR Plus 2019; 3:e10177. [PMID: 30828691 PMCID: PMC6383702 DOI: 10.1002/jbm4.10177] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/29/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022] Open
Abstract
Recent studies using genetically modified mice, pharmacological approaches, and human samples have highlighted an important role for the epidermal growth factor receptor (EGFR), selected ligands, and downstream components in endochondral bone formation and joint homeostasis. Although most data demonstrate an important function of this pathway in endochondral ossification and articular cartilage growth, conflicting results on its role in osteoarthritis have been reported. In some contexts, inactivation of EGFR signaling has been shown to protect joints from surgically induced osteoarthritis, whereas in others, similar manipulations worsened joint pathology. The current review summarizes recent studies of cartilage EGFR signaling in long bone development and diseases, provides potential explanations for the reported discrepancies, and suggests directions for future work to clarify the potential of this pathway as target for osteoarthritis treatment. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ling Qin
- Department of Orthopaedic SurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Frank Beier
- Department of Physiology and PharmacologyUniversity of Western OntarioLondonCanada
- Western Bone and Joint InstituteUniversity of Western OntarioLondonCanada
| |
Collapse
|
12
|
Araya HF, Sepulveda H, Lizama CO, Vega OA, Jerez S, Briceño PF, Thaler R, Riester SM, Antonelli M, Salazar-Onfray F, Rodríguez JP, Moreno RD, Montecino M, Charbonneau M, Dubois CM, Stein GS, van Wijnen AJ, Galindo MA. Expression of the ectodomain-releasing protease ADAM17 is directly regulated by the osteosarcoma and bone-related transcription factor RUNX2. J Cell Biochem 2018; 119:8204-8219. [PMID: 29923217 DOI: 10.1002/jcb.26832] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/06/2018] [Indexed: 01/04/2023]
Abstract
Osteoblast differentiation is controlled by transcription factor RUNX2 which temporally activates or represses several bone-related genes, including those encoding extracellular matrix proteins or factors that control cell-cell, and cell-matrix interactions. Cell-cell communication in the many skeletal pericellular micro-niches is critical for bone development and involves paracrine secretion of growth factors and morphogens. This paracrine signaling is in part regulated by "A Disintegrin And Metalloproteinase" (ADAM) proteins. These cell membrane-associated metalloproteinases support proteolytic release ("shedding") of protein ectodomains residing at the cell surface. We analyzed microarray and RNA-sequencing data for Adam genes and show that Adam17, Adam10, and Adam9 are stimulated during BMP2 mediated induction of osteogenic differentiation and are robustly expressed in human osteoblastic cells. ADAM17, which was initially identified as a tumor necrosis factor alpha (TNFα) converting enzyme also called (TACE), regulates TNFα-signaling pathway, which inhibits osteoblast differentiation. We demonstrate that Adam17 expression is suppressed by RUNX2 during osteoblast differentiation through the proximal Adam17 promoter region (-0.4 kb) containing two functional RUNX2 binding motifs. Adam17 downregulation during osteoblast differentiation is paralleled by increased RUNX2 expression, cytoplasmic-nuclear translocation and enhanced binding to the Adam17 proximal promoter. Forced expression of Adam17 reduces Runx2 and Alpl expression, indicating that Adam17 may negatively modulate osteoblast differentiation. These findings suggest a novel regulatory mechanism involving a reciprocal Runx2-Adam17 negative feedback loop to regulate progression through osteoblast differentiation. Our results suggest that RUNX2 may control paracrine signaling through regulation of ectodomain shedding at the cell surface of osteoblasts by directly suppressing Adam17 expression.
Collapse
Affiliation(s)
- Héctor F Araya
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Hugo Sepulveda
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Carlos O Lizama
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar A Vega
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sofia Jerez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pedro F Briceño
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Marcelo Antonelli
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Pablo Rodríguez
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), University of Chile, Santiago, Chile
| | - Ricardo D Moreno
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Martin Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Martine Charbonneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Claire M Dubois
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, The Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mario A Galindo
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
13
|
Yang CY, Chanalaris A, Troeberg L. ADAMTS and ADAM metalloproteinases in osteoarthritis - looking beyond the 'usual suspects'. Osteoarthritis Cartilage 2017; 25:1000-1009. [PMID: 28216310 PMCID: PMC5473942 DOI: 10.1016/j.joca.2017.02.791] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Matrix metalloproteinases (MMPs) and 'aggrecanase' a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) are well established to play key roles in osteoarthritis (OA) through degradation of extracellular matrix (ECM) type II collagen and aggrecan, and are thus potential targets for development of OA therapies. OBJECTIVE This paper aims to provide a comprehensive review of the expression and potential roles of other, lesser-known ADAMTSs and related adamalysins (or a disintegrin and metalloproteinases (ADAMs)) in cartilage, with a view to identifying potentially protective or homeostatic metalloproteinases in the joint and informing consequent selective inhibitor design. DESIGN A comprehensive literature search was performed using PubMed terms 'osteoarthritis' and 'ADAMTS' or 'ADAM'. RESULTS Several ADAMTSs and ADAMs were identified as having reportedly increased expression in OA. These include enzymes likely to play roles in cartilage matrix anabolism (e.g., the procollagen N-proteinases ADAMTS-2, ADAMTS-3 and ADAMTS-14), chondrocyte differentiation and proliferation (e.g., ADAM9, ADAM10, ADAM12), as well as enzymes contributing to cartilage catabolism (e.g., Cartilage oligomeric protein (COMP)-degrading ADAMTS-7 and ADAMTS-12). CONCLUSIONS In addition to the well-characterised MMPs, ADAMTS-4 and ADAMTS-5, many other ADAMTSs and ADAMs are expressed in cartilage and several show significantly altered expression in OA. Studies aimed at elucidating the pathophysiological roles of these enzymes in cartilage will contribute to our understanding of OA pathogenesis and enable design of targeted inhibitors that effectively target metalloproteinase-mediated cartilage degradation while sparing cartilage repair pathways.
Collapse
Affiliation(s)
| | | | - L. Troeberg
- Address correspondence and reprint requests to: L. Troeberg, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, OX3 7FY Oxford, UK.Kennedy Institute of RheumatologyUniversity of OxfordRoosevelt DriveOxfordOX3 7FYUK
| |
Collapse
|
14
|
Mizuno S, Yoda M, Shimoda M, Tohmonda T, Okada Y, Toyama Y, Takeda S, Nakamura M, Matsumoto M, Horiuchi K. A Disintegrin and Metalloprotease 10 (ADAM10) Is Indispensable for Maintenance of the Muscle Satellite Cell Pool. J Biol Chem 2015; 290:28456-28464. [PMID: 26453297 DOI: 10.1074/jbc.m115.653477] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Indexed: 12/20/2022] Open
Abstract
Satellite cells (SCs) are muscle-specific stem cells that are essential for the regeneration of damaged muscles. Although SCs have a robust capacity to regenerate myofibers, the number of SCs decreases with aging, leading to insufficient recovery after muscle injury. We herein show that ADAM10 (a disintegrin and metalloprotease 10), a membrane-bound proteolytic enzyme with a critical role in Notch processing (S2 cleavage), is essential for the maintenance of SC quiescence. We generated mutant mice in which ADAM10 in SCs can be conditionally abrogated by tamoxifen injection. Tamoxifen-treated mutant mice did not show any apparent defects and grew normally under unchallenged conditions. However, these mice showed a nearly complete loss of muscle regeneration after chemically induced muscle injury. In situ hybridization and flow cytometric analyses revealed that the mutant mice had significantly less SCs compared with wild type controls. Of note, we found that inactivation of ADAM10 in SCs severely compromised Notch signaling and led to dysregulated myogenic differentiation, ultimately resulting in deprivation of the SC pool in vivo. Taken together, the present findings underscore the role of ADAM10 as an indispensable component of Notch signaling in SCs and for maintaining the SC pool.
Collapse
Affiliation(s)
- Sakiko Mizuno
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masaki Yoda
- Anti-aging Orthopedic Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masayuki Shimoda
- Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takahide Tohmonda
- Anti-aging Orthopedic Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasunori Okada
- Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yoshiaki Toyama
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Masaya Nakamura
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Morio Matsumoto
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keisuke Horiuchi
- Departments of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan; Anti-aging Orthopedic Research, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
15
|
Secreted Frizzled-related protein 3 (sFRP3)-mediated suppression of interleukin-6 receptor release by A disintegrin and metalloprotease 17 (ADAM17) is abrogated in the osteoarthritis-associated rare double variant of sFRP3. Biochem J 2015; 468:507-18. [DOI: 10.1042/bj20141231] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 04/07/2015] [Indexed: 11/17/2022]
Abstract
A disintegrin and metalloprotease 17 (ADAM17) activity and secreted Frizzled-related protein 3 (sFRP3) down-regulation or expression of its rare double variant is associated with arthritis. sFRP3 interacts with interleukin-6 receptor (IL-6R) and ADAM17 and suppresses ADAM17 activity, whereas the rare variant does not; these findings provide explanation for their opposing pathogenic associations.
Collapse
|
16
|
Sun MMG, Beier F. Chondrocyte hypertrophy in skeletal development, growth, and disease. ACTA ACUST UNITED AC 2015; 102:74-82. [PMID: 24677724 DOI: 10.1002/bdrc.21062] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 02/27/2014] [Indexed: 12/31/2022]
Abstract
Most of our bones form through the process of endochondral ossification, which is tightly regulated by the activity of the cartilage growth plate. Chondrocyte maturation through the various stages of growth plate physiology ultimately results in hypertrophy. Chondrocyte hypertrophy is an essential contributor to longitudinal bone growth, but recent data suggest that these cells also play fundamental roles in signaling to other skeletal cells, thus coordinating endochondral ossification. On the other hand, ectopic hypertrophy of articular chondrocytes has been implicated in the pathogenesis of osteoarthritis. Thus, a better understanding of the processes that control chondrocyte hypertrophy in the growth plate as well as in articular cartilage is required for improved management of both skeletal growth disorders and osteoarthritis. This review summarizes recent findings on the regulation of hypertrophic chondrocyte differentiation, the cellular mechanisms involved in hypertrophy, and the role of chondrocyte hypertrophy in skeletal physiology and pathophysiology.
Collapse
Affiliation(s)
- Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Western University, and Children's Health Research Institute, London, Ontario, Canada
| | | |
Collapse
|
17
|
Chen Z, Yue SX, Zhou G, Greenfield EM, Murakami S. ERK1 and ERK2 regulate chondrocyte terminal differentiation during endochondral bone formation. J Bone Miner Res 2015; 30:765-74. [PMID: 25401279 PMCID: PMC4487783 DOI: 10.1002/jbmr.2409] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 11/06/2014] [Accepted: 11/12/2014] [Indexed: 12/29/2022]
Abstract
Chondrocytes in the epiphyseal cartilage undergo terminal differentiation prior to their removal through apoptosis. To examine the role of ERK1 and ERK2 in chondrocyte terminal differentiation, we generated Osterix (Osx)-Cre; ERK1(-/-) ; ERK2(flox/flox) mice (conditional knockout Osx [cKOosx]), in which ERK1 and ERK2 were deleted in hypertrophic chondrocytes. These cKOosx mice were grossly normal in size at birth, but by 3 weeks of age exhibited shorter long bones. Histological analysis in these mice revealed that the zone of hypertrophic chondrocytes in the growth plate was markedly expanded. In situ hybridization and quantitative real-time PCR analyses demonstrated that Matrix metalloproteinase-13 (Mmp13) and Osteopontin expression was significantly decreased, indicating impaired chondrocyte terminal differentiation. Moreover, Egr1 and Egr2, transcription factors whose expression is restricted to the last layers of hypertrophic chondrocytes in wild-type mice, were also strongly downregulated in these cKOosx mice. In transient transfection experiments in the RCS rat chondrosarcoma cell line, the expression of Egr1, Egr2, or a constitutively active mutant of MEK1 increased the activity of an Osteopontin promoter, whereas the MEK1-induced activation of the Osteopontin promoter was inhibited by the coexpression of Nab2, an Egr1 and Egr2 co-repressor. These results suggest that MEK1-ERK signaling activates the Osteopontin promoter in part through Egr1 and Egr2. Finally, our histological analysis of cKOosx mice demonstrated enchondroma-like lesions in the bone marrow that are reminiscent of human metachondromatosis, a skeletal disorder caused by mutations in PTPN11. Our observations suggest that the development of enchondromas in metachondromatosis may be caused by reduced extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK MAPK) signaling.
Collapse
Affiliation(s)
- Zhijun Chen
- Department of Orthopaedics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Susan X. Yue
- Department of Orthopaedics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Guang Zhou
- Department of Orthopaedics, Case Western Reserve University, Cleveland, Ohio 44106
- Department of Genetics and Genomic Sciences, Case Western Reserve University, Cleveland, Ohio 44106
| | - Edward M. Greenfield
- Department of Orthopaedics, Case Western Reserve University, Cleveland, Ohio 44106
- Division of General Medical Sciences, National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Shunichi Murakami
- Department of Orthopaedics, Case Western Reserve University, Cleveland, Ohio 44106
- Department of Genetics and Genomic Sciences, Case Western Reserve University, Cleveland, Ohio 44106
- Division of General Medical Sciences, National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio 44106
- Murakami Geka Iin, Kawasaki, 210-0834 Japan
- Corresponding author: Shunichi Murakami, 11100 Euclid Avenue, Hanna House 6th floor, Cleveland, Ohio 44106, phone: 216-368-3965, fax: 216-368-1332,
| |
Collapse
|
18
|
Pest MA, Russell BA, Zhang YW, Jeong JW, Beier F. Disturbed cartilage and joint homeostasis resulting from a loss of mitogen-inducible gene 6 in a mouse model of joint dysfunction. Arthritis Rheumatol 2014; 66:2816-27. [PMID: 24966136 DOI: 10.1002/art.38758] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 06/19/2014] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Mitogen-inducible gene 6 (MIG-6) regulates epidermal growth factor receptor (EGFR) signaling in synovial joint tissues. Whole-body knockout of the Mig6 gene in mice has been shown to induce osteoarthritis and joint degeneration. To evaluate the role of chondrocytes in this process, Mig6 was conditionally deleted from Col2a1-expressing cell types in the cartilage of mice. METHODS Bone and cartilage in the synovial joints of cartilage-specific Mig6-deleted (knockout [KO]) mice and control littermates were compared. Histologic staining and immunohistochemical analyses were used to evaluate joint pathology as well as the expression of key extracellular matrix and regulatory proteins. Calcified tissue in synovial joints was assessed by micro-computed tomography (micro-CT) and whole-skeleton staining. RESULTS Formation of long bones was found to be normal in KO animals. Cartilage thickness and proteoglycan staining of articular cartilage in the knee joints of 12-week-old KO mice were increased as compared to controls, with higher cellularity throughout the tissue. Radiopaque chondro-osseous nodules appeared in the knees of KO animals by 12 weeks of age and progressed to calcified bone-like tissue by 36 weeks of age. Nodules were also observed in the spine of 36-week-old animals. Erosion of bone at ligament entheses was evident by 12 weeks of age, by both histologic and micro-CT assessment. CONCLUSION MIG-6 expression in chondrocytes is important for the maintenance of cartilage and joint homeostasis. Dysregulation of EGFR signaling in chondrocytes results in anabolic activity in cartilage, but erosion of ligament entheses and the formation of ectopic chondro-osseous nodules severely disturb joint physiology.
Collapse
|
19
|
Zhang X, Zhu J, Liu F, Li Y, Chandra A, Levin LS, Beier F, Enomoto-Iwamoto M, Qin L. Reduced EGFR signaling enhances cartilage destruction in a mouse osteoarthritis model. Bone Res 2014; 2:14015. [PMID: 26120493 PMCID: PMC4472123 DOI: 10.1038/boneres.2014.15] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 11/12/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease
and a major cause of pain and disability in older
adults. We have previously identified epidermal growth
factor receptor (EGFR) signaling as an
important regulator of cartilage matrix degradation
during epiphyseal cartilage development. To study its
function in OA progression, we performed surgical
destabilization of the medial meniscus (DMM)
to induce OA in two mouse models with reduced EGFR
activity, one with genetic modification
(EgfrWa5/+
mice) and the other one with pharmacological
inhibition (gefitinib treatment).
Histological analyses and scoring at 3 months
post-surgery revealed increased cartilage destruction
and accelerated OA progression in both mouse models.
TUNEL staining demonstrated that EGFR signaling
protects chondrocytes from OA-induced apoptosis, which
was further confirmed in primary chondrocyte culture.
Immunohistochemistry showed increased aggrecan
degradation in these mouse models, which coincides with
elevated amounts of ADAMTS5 and matrix
metalloproteinase 13 (MMP13), the principle
proteinases responsible for aggrecan degradation, in
the articular cartilage after DMM surgery. Furthermore,
hypoxia-inducible factor 2α
(HIF2α), a critical catabolic
transcription factor stimulating MMP13 expression
during OA, was also upregulated in mice with reduced
EGFR signaling. Taken together, our findings
demonstrate a primarily protective role of EGFR during
OA progression by regulating chondrocyte survival and
cartilage degradation.
Collapse
Affiliation(s)
- Xianrong Zhang
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA ; Department of Physiology, School of Basic Medical Sciences, Wuhan University , Wuhan, China
| | - Ji Zhu
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - Fei Liu
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA ; Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital , Shanghai, China
| | - Yumei Li
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA ; Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Abhishek Chandra
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - L Scott Levin
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, ON, Canada
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA ; Department of Surgery, The Children's Hospital of Philadelphia , Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, School of Medicine, University of Pennsylvania , Philadelphia, PA, USA
| |
Collapse
|
20
|
Developmental defects in zebrafish for classification of EGF pathway inhibitors. Toxicol Appl Pharmacol 2013; 274:339-49. [PMID: 24262764 DOI: 10.1016/j.taap.2013.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 01/22/2023]
Abstract
One of the major challenges when testing drug candidates targeted at a specific pathway in whole animals is the discrimination between specific effects and unwanted, off-target effects. Here we used the zebrafish to define several developmental defects caused by impairment of Egf signaling, a major pathway of interest in tumor biology. We inactivated Egf signaling by genetically blocking Egf expression or using specific inhibitors of the Egf receptor function. We show that the combined occurrence of defects in cartilage formation, disturbance of blood flow in the trunk and a decrease of myelin basic protein expression represent good indicators for impairment of Egf signaling. Finally, we present a classification of known tyrosine kinase inhibitors according to their specificity for the Egf pathway. In conclusion, we show that developmental indicators can help to discriminate between specific effects on the target pathway from off-target effects in molecularly targeted drug screening experiments in whole animal systems.
Collapse
|
21
|
ADAM17 controls endochondral ossification by regulating terminal differentiation of chondrocytes. Mol Cell Biol 2013. [PMID: 23732913 DOI: 10.1128/mcb.00291‐13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Endochondral ossification is a highly regulated process that relies on properly orchestrated cell-cell interactions in the developing growth plate. This study is focused on understanding the role of a crucial regulator of cell-cell interactions, the membrane-anchored metalloproteinase ADAM17, in endochondral ossification. ADAM17 releases growth factors, cytokines, and other membrane proteins from cells and is essential for epidermal growth factor receptor (EGFR) signaling and for processing tumor necrosis factor alpha. Here, we report that mice lacking ADAM17 in chondrocytes (A17ΔCh) have a significantly expanded zone of hypertrophic chondrocytes in the growth plate and retarded growth of long bones. This abnormality is caused by an accumulation of the most terminally differentiated type of chondrocytes that produces a calcified matrix. Inactivation of ADAM17 in osteoclasts or endothelial cells does not affect the zone of hypertrophic chondrocytes, suggesting that the main role of ADAM17 in the growth plate is in chondrocytes. This notion is further supported by in vitro experiments showing enhanced hypertrophic differentiation of primary chondrocytes lacking Adam17. The enlarged zone of hypertrophic chondrocytes in A17ΔCh mice resembles that described in mice with mutant EGFR signaling or lack of its ligand transforming growth factor α (TGFα), suggesting that ADAM17 regulates terminal differentiation of chondrocytes during endochondral ossification by activating the TGFα/EGFR signaling axis.
Collapse
|
22
|
ADAM17 controls endochondral ossification by regulating terminal differentiation of chondrocytes. Mol Cell Biol 2013; 33:3077-90. [PMID: 23732913 DOI: 10.1128/mcb.00291-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Endochondral ossification is a highly regulated process that relies on properly orchestrated cell-cell interactions in the developing growth plate. This study is focused on understanding the role of a crucial regulator of cell-cell interactions, the membrane-anchored metalloproteinase ADAM17, in endochondral ossification. ADAM17 releases growth factors, cytokines, and other membrane proteins from cells and is essential for epidermal growth factor receptor (EGFR) signaling and for processing tumor necrosis factor alpha. Here, we report that mice lacking ADAM17 in chondrocytes (A17ΔCh) have a significantly expanded zone of hypertrophic chondrocytes in the growth plate and retarded growth of long bones. This abnormality is caused by an accumulation of the most terminally differentiated type of chondrocytes that produces a calcified matrix. Inactivation of ADAM17 in osteoclasts or endothelial cells does not affect the zone of hypertrophic chondrocytes, suggesting that the main role of ADAM17 in the growth plate is in chondrocytes. This notion is further supported by in vitro experiments showing enhanced hypertrophic differentiation of primary chondrocytes lacking Adam17. The enlarged zone of hypertrophic chondrocytes in A17ΔCh mice resembles that described in mice with mutant EGFR signaling or lack of its ligand transforming growth factor α (TGFα), suggesting that ADAM17 regulates terminal differentiation of chondrocytes during endochondral ossification by activating the TGFα/EGFR signaling axis.
Collapse
|