1
|
Zhang L, Zhou J, Kong W. Extracellular matrix in vascular homeostasis and disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01103-0. [PMID: 39743560 DOI: 10.1038/s41569-024-01103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 01/04/2025]
Abstract
The extracellular matrix is an essential component and constitutes a dynamic microenvironment of the vessel wall with an indispensable role in vascular homeostasis and disease. From early development through to ageing, the vascular extracellular matrix undergoes various biochemical and biomechanical alterations in response to diverse environmental cues and exerts precise regulatory control over vessel remodelling. Advances in novel technologies that enable the comprehensive evaluation of extracellular matrix components and cell-matrix interactions have led to the emergence of therapeutic strategies that specifically target this fine-tuned network. In this Review, we explore various aspects of extracellular matrix biology in vascular development, disorders and ageing, emphasizing the effect of the extracellular matrix on disease initiation and progression. Additionally, we provide an overview of the potential therapeutic implications of targeting the extracellular matrix microenvironment in vascular diseases.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
2
|
Gao R, Hu Y, Yuan Q. ADAMTS12 serves as a novel prognostic biomarker and promotes proliferation and invasion in gastric cancer. Discov Oncol 2024; 15:837. [PMID: 39720953 DOI: 10.1007/s12672-024-01724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Gastric cancer (GC) remains a prevalent and aggressive malignancy with a poor prognosis. This study aimed to identify diagnostic and prognostic biomarkers while exploring their potential functions in GC. A total of 598 upregulated and 506 downregulated genes were identified in GC patients. Among these, survival-related differentially expressed genes (DEGs), including ADAMTS12, F5, and VCAN, were highlighted. Pan-cancer analyses revealed their dysregulation across multiple tumor types. A novel prognostic signature, incorporating ADAMTS12 and F5, effectively stratified GC patients into low- and high-risk groups, demonstrating significant differences in overall survival and robust predictive performance. ADAMTS12, strongly associated with advanced clinical stages and poor prognosis, was validated in an independent cohort and exhibited promising diagnostic potential. RT-PCR and western blot analyses confirmed its high expression in GC tissues and cell lines. Functional assays further demonstrated that ADAMTS12 promotes GC cell proliferation and invasion. In summary, this study provides critical insights into the molecular landscape of GC, offering a potential prognostic tool and therapeutic target.
Collapse
Affiliation(s)
- Ruimei Gao
- Department of Gastroenterology, Qingdao Chengyang People's Hospital, Qingdao, China
| | - Yalan Hu
- Department of Anorectal Surgery, Qingdao Eighth People's Hospital, Qingdao, China
| | - Qiuxiang Yuan
- Department of Gastroenterology, Qingdao Chengyang People's Hospital, Qingdao, China.
| |
Collapse
|
3
|
Larsson S, Lalande A, Stefan Lohmander L, Soret P, Bernard K, Pueyo M, Struglics A. Serum ARGS-aggrecan in a phase 2 clinical trial targeting osteoarthritis. Osteoarthritis Cartilage 2024; 32:1463-1470. [PMID: 38862084 DOI: 10.1016/j.joca.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To monitor serum concentrations of the aggrecan alanine-arginine-glycine-serine (ARGS) neoepitope in a clinical trial of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-5 inhibition as disease-modifying therapy of knee osteoarthritis, and to investigate relationships between reduction in ARGS and change in cartilage thickness, knee-related pain and function. DESIGN ROCCELLA trial participants received once-daily oral S201086 75, 150 or 300 mg, or placebo, for 52 weeks. Serum was collected at baseline, 4, 12, 28 and 52 weeks, and 2 weeks post-treatment with ARGS measured by an in-house immunoassay. Change from baseline to week 52 in central medial femorotibial compartment cartilage thickness was measured by magnetic resonance imaging, function and pain by Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) subscores. Associations between cumulative change in ARGS and change in cartilage thickness or WOMAC subscores were evaluated by linear regression. RESULTS S201086 reduced serum levels of ARGS in a dose-dependent manner throughout the treatment period. Maximal reduction was at 4 weeks with a 58.5% [95% CI 60.8%, 56.2%] reduction of ARGS compared to baseline for 300 mg S201086. Two weeks post-treatment, ARGS concentrations rebounded with a dose-dependent overshoot compared to baseline levels. Cumulative change of ARGS concentration from baseline to week 52 had no effect on change in cartilage thickness (slope -0.8×10-6 [-2.9×10-6, 1.3×10-6]) or change in WOMAC pain and function (slopes -30×10-6 [-64×10-6, 5.2×10-6] and -97×10-6 [-214×10-6, 20×10-6], respectively) at week 52. CONCLUSION Systemic inhibition of ADAMTS-5 resulted in markedly reduced serum ARGS, but change in serum ARGS concentrations showed no association with the progression of cartilage thinning, or patient reported pain and function.
Collapse
Affiliation(s)
- Staffan Larsson
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| | - Agnès Lalande
- Institut de Recherches et Développement Servier Paris SACLAY, Gif-sur-Yvette, France.
| | - L Stefan Lohmander
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| | - Perrine Soret
- Institut de Recherches et Développement Servier Paris SACLAY, Gif-sur-Yvette, France.
| | - Katy Bernard
- Institut de Recherches et Développement Servier Paris SACLAY, Gif-sur-Yvette, France.
| | - Maria Pueyo
- Institut de Recherches et Développement Servier Paris SACLAY, Gif-sur-Yvette, France.
| | - André Struglics
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| |
Collapse
|
4
|
Mead TJ, Bhutada S, Foulcer SJ, Peruzzi N, Nelson CM, Seifert DE, Larkin J, Tran-Lundmark K, Filmus J, Apte SS. Combined genetic-pharmacologic inactivation of tightly linked ADAMTS proteases in temporally specific windows uncovers distinct roles for versican proteolysis and glypican-6 in cardiac development. Matrix Biol 2024; 131:1-16. [PMID: 38750698 PMCID: PMC11526477 DOI: 10.1016/j.matbio.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Extracellular matrix remodeling mechanisms are understudied in cardiac development and congenital heart defects. We show that matrix-degrading metalloproteases ADAMTS1 and ADAMTS5, are extensively co-expressed during mouse cardiac development. The mouse mutants of each gene have mild cardiac anomalies, however, their combined genetic inactivation to elicit cooperative roles is precluded by tight gene linkage. Therefore, we coupled Adamts1 inactivation with pharmacologic ADAMTS5 blockade to uncover stage-specific cooperative roles and investigated their potential substrates in mouse cardiac development. ADAMTS5 blockade was achieved in Adamts1 null mouse embryos using an activity-blocking monoclonal antibody during distinct developmental windows spanning myocardial compaction or cardiac septation and outflow tract rotation. Synchrotron imaging, RNA in situ hybridization, immunofluorescence microscopy and electron microscopy were used to determine the impact on cardiac development and compared to Gpc6 and ADAMTS-cleavage resistant versican mutants. Mass spectrometry-based N-terminomics was used to seek relevant substrates. Combined inactivation of ADAMTS1 and ADAMTS5 prior to 12.5 days of gestation led to dramatic accumulation of versican-rich cardiac jelly and inhibited formation of compact and trabecular myocardium, which was also observed in mice with ADAMTS cleavage-resistant versican. Combined inactivation after 12.5 days impaired outflow tract development and ventricular septal closure, generating a tetralogy of Fallot-like defect. N-terminomics of combined ADAMTS knockout and control hearts identified a cleaved glypican-6 peptide only in the controls. ADAMTS1 and ADAMTS5 expression in cells was associated with specific glypican-6 cleavages. Paradoxically, combined ADAMTS1 and ADAMTS5 inactivation reduced cardiac glypican-6 and outflow tract Gpc6 transcription. Notably, Gpc6-/- hearts demonstrated similar rotational defects as combined ADAMTS inactivated hearts and both had reduced hedgehog signaling. Thus, versican proteolysis in cardiac jelly at the canonical Glu441-Ala442 site is cooperatively mediated by ADAMTS1 and ADAMTS5 and required for proper ventricular cardiomyogenesis, whereas, reduced glypican-6 after combined ADAMTS inactivation impairs hedgehog signaling, leading to outflow tract malrotation.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA; University Hospitals Rainbow Babies and Children's Hospital, Cleveland, OH, USA.
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Simon J Foulcer
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Niccolò Peruzzi
- Department of Experimental Medical Science, and Wallenberg Center for Molecular Medicine Lund University and The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Courtney M Nelson
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Deborah E Seifert
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Karin Tran-Lundmark
- Department of Experimental Medical Science, and Wallenberg Center for Molecular Medicine Lund University and The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Jorge Filmus
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
5
|
Sun GB, Lu YF, Duan XJ. Investigation into the association of FNDC1 and ADAMTS12 gene expression with plumage coloration in Muscovy ducks. Open Life Sci 2024; 19:20220877. [PMID: 38867923 PMCID: PMC11167702 DOI: 10.1515/biol-2022-0877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024] Open
Abstract
To elucidate the molecular genetic mechanisms underpinning feather color in Muscovy ducks. A cohort of 100 Muscovy ducks was meticulously selected for this research. Follicular tissues from ducks exhibiting black and white plumage served as the experimental samples. From these tissues, RNA and proteins were extracted for further analysis. The RNA underwent reverse transcription polymerase chain reaction amplification, followed by validation through western blot assays. The data revealed a significant upregulation in the expression of FN domain-containing protein 1 (FNDC1) and ADAMTS12 genes in Muscovy ducks with white plumage traits as opposed to those with black plumage traits. Specifically, individuals with pure white plumage demonstrated a markedly elevated expression of the FNDC1 gene in comparison to their pure black counterparts. Conversely, expression levels of the ADAMTS12 gene were found to be reduced in ducks with pure black plumage relative to those with pure white plumage. Notably, the expression patterns of FNDC1 and ADAMTS12 genes exhibited inconsistencies between mRNA and protein levels. This study offers significant insights into the molecular genetic mechanisms underlying feather color variation in Muscovy ducks. FNDC1 and ADAMTS12 could be considered potential targets for genetic manipulation or selective breeding strategies aimed at achieving specific feather color phenotypes in Muscovy ducks.
Collapse
Affiliation(s)
- Guo-Bo Sun
- Animal Science and Technology College, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu, China
| | - Yan-Feng Lu
- Animal Science and Technology College, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu, China
| | - Xiu-Jun Duan
- Animal Science and Technology College, Jiangsu Agri-animal Husbandry Vocational College, No. 8 of Fenghuang East Road, Hailing District, Taizhou, 225300, Jiangsu, China
| |
Collapse
|
6
|
Sarıkaya B, Dolap MA, Kaptan AY, Bozkurt C, Yumuşak N, Yigin A, Sipahioğlu S, Çetin BV, Altay MA. Histological Structure and Immunohistochemical Properties of the Ligamentum Teres in Patients With Developmental Dysplasia of the Hip. Cureus 2024; 16:e59748. [PMID: 38841047 PMCID: PMC11152455 DOI: 10.7759/cureus.59748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction This study aims to evaluate the histology of the ligamentum teres and its relationship with matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), which are involved in the destruction of extracellular matrix proteins in patients with developmental dysplasia of the hip (DDH). Methodology The patients who underwent open reduction and pelvic osteotomy due to DDH were included in the study. Patient groups were formed according to Tönnis stages, positive family history, consanguineous marriage, age, and bilateral involvement. The histology and immunohistochemical properties (MMP-2, MMP-9, and ADAMTS-7) of ligamentum teres tissue obtained from the patients were evaluated according to these groups. Results Thirty-five patients (female 30, 85.7%; male 5, 14.3%) with DDH between the ages of 14 and 99 months were included in the study. Preoperative and postoperative Tönnis stages, positive family history, consanguineous marriage, age, and bilaterality did not cause a significant difference between histological parameters. A significant correlation was found between MMP-2, MMP-9, and ADAMTS-7 and all histological parameters. Conclusions The histological structure of ligamentum teres in patients with DDH shows moderate inflammation, fibrosis, neovascularization, hyalinization, and fatty infiltration regardless of age and radiological stage. ADAMTS-7, MMP-2, and MMP-9 correlate positively with the histological parameters of the ligamentum teres in patients with DDH.
Collapse
Affiliation(s)
- Baran Sarıkaya
- Department of Orthopaedics and Traumatology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, TUR
| | - Mehmet Ali Dolap
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Harran University, Sanliurfa, TUR
| | - Ahmet Yiğit Kaptan
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Harran University, Sanliurfa, TUR
| | - Celal Bozkurt
- Department of Orthopaedics and Traumatology, Gaziosmanpaşa Taksim Training and Research Hospital, University of Health Sciences, Istanbul, TUR
| | - Nihat Yumuşak
- Department of Pathology, Faculty of Veterinary, Harran University, Sanliurfa, TUR
| | - Akin Yigin
- Department of Genetics, Faculty of Veterinary, Harran University, Sanliurfa, TUR
| | - Serkan Sipahioğlu
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Ordu University, Ordu, TUR
| | - Baki Volkan Çetin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Harran University, Sanliurfa, TUR
| | - Mehmet Akif Altay
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Harran University, Sanliurfa, TUR
| |
Collapse
|
7
|
Meibom D, Wasnaire P, Beyer K, Broehl A, Cancho-Grande Y, Elowe N, Henninger K, Johannes S, Jungmann N, Krainz T, Lindner N, Maassen S, MacDonald B, Menshykau D, Mittendorf J, Sanchez G, Schaefer M, Stefan E, Torge A, Xing Y, Zubov D. BAY-9835: Discovery of the First Orally Bioavailable ADAMTS7 Inhibitor. J Med Chem 2024; 67:2907-2940. [PMID: 38348661 PMCID: PMC10895658 DOI: 10.1021/acs.jmedchem.3c02036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/23/2024]
Abstract
The matrix metalloprotease ADAMTS7 has been identified by multiple genome-wide association studies as being involved in the development of coronary artery disease. Subsequent research revealed the proteolytic function of the enzyme to be relevant for atherogenesis and restenosis after vessel injury. Based on a publicly known dual ADAMTS4/ADAMTS5 inhibitor, we have in silico designed an ADAMTS7 inhibitor of the catalytic domain, which served as a starting point for an optimization campaign. Initially our inhibitors suffered from low selectivity vs MMP12. An X-ray cocrystal structure inspired us to exploit amino acid differences in the binding site of MMP12 and ADAMTS7 to improve selectivity. Further optimization composed of employing 5-membered heteroaromatic groups as hydantoin substituents to become more potent on ADAMTS7. Finally, fine-tuning of DMPK properties yielded BAY-9835, the first orally bioavailable ADAMTS7 inhibitor. Further optimization to improve selectivity vs ADAMTS12 seems possible, and a respective starting point could be identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eric Stefan
- Broad
Institute, 02142 Cambridge, United States
| | | | - Yi Xing
- Broad
Institute, 02142 Cambridge, United States
| | | |
Collapse
|
8
|
Mead TJ, Bhutada S, Martin DR, Apte SS. Proteolysis: a key post-translational modification regulating proteoglycans. Am J Physiol Cell Physiol 2022; 323:C651-C665. [PMID: 35785985 PMCID: PMC9448339 DOI: 10.1152/ajpcell.00215.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022]
Abstract
Proteoglycans are composite molecules comprising a protein backbone, i.e., the core protein, with covalently attached glycosaminoglycan chains of distinct chemical types. Most proteoglycans are secreted or attached to the cell membrane. Their specialized structures, binding properties, and biophysical attributes underlie diverse biological roles, which include modulation of tissue mechanics, cell adhesion, and the sequestration and regulated release of morphogens, growth factors, and cytokines. As an irreversible post-translational modification, proteolysis has a profound impact on proteoglycan function, abundance, and localization. Proteolysis is required for molecular maturation of some proteoglycans, clearance of extracellular matrix proteoglycans during tissue remodeling, generation of bioactive fragments from proteoglycans, and ectodomain shedding of cell-surface proteoglycans. Genetic evidence shows that proteoglycan core protein proteolysis is essential for diverse morphogenetic events during embryonic development. In contrast, dysregulated proteoglycan proteolysis contributes to osteoarthritis, cardiovascular disorders, cancer, and inflammation. Proteolytic fragments of perlecan, versican, aggrecan, brevican, collagen XVIII, and other proteoglycans are associated with independent biological activities as so-called matrikines. Yet, proteoglycan proteolysis has been investigated to only a limited extent to date. Here, we review the actions of proteases on proteoglycans and illustrate their functional impact with several examples. We discuss the applications and limitations of strategies used to define cleavage sites in proteoglycans and explain how proteoglycanome-wide proteolytic mapping, which is desirable to fully understand the impact of proteolysis on proteoglycans, can be facilitated by integrating classical proteoglycan isolation methods with mass spectrometry-based proteomics.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Daniel R Martin
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
9
|
Dudiki T, Nascimento DW, Childs LS, Kareti S, Androjna C, Zhevlakova I, Byzova TV. Progressive skeletal defects caused by Kindlin3 deficiency, a model of autosomal recessive osteopetrosis in humans. Bone 2022; 160:116397. [PMID: 35342016 PMCID: PMC9133165 DOI: 10.1016/j.bone.2022.116397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/18/2022] [Accepted: 03/21/2022] [Indexed: 11/20/2022]
Abstract
The cellular and molecular mechanisms of bone development and homeostasis are clinically important, but not fully understood. Mutations in integrins and Kindlin3 in humans known as Leukocyte adhesion deficiencies (LAD) cause a wide spectrum of complications, including osteopetrosis. Yet, the rarity, frequent misdiagnosis, and lethality of LAD preclude mechanistic analysis of skeletal abnormalities in these patients. Here, using inducible and constitutive tissue-specific Kindlin3 knockout (K3KO) mice, we show that the constitutive lack of embryonic-Kindlin3 in myeloid lineage cells causes growth retardation, edentulism, and skull deformity indicative of hydrocephaly. Micro-CT analysis revealed craniosynostosis, choanal stenosis, and micrognathia along with other skeletal abnormalities characteristic of osteopetrosis. A marked progression of osteosclerosis occurs in mature to middle-aged adults, resulting in the narrowing of cranial nerve foramina and bone marrow cavities of long bones. However, postnatal-Kindlin3 is less critical for bone remodeling and architecture. Thus, myeloid Kindlin3 is essential for skeletal development and its deficiency leads to autosomal recessive osteopetrosis (ARO). The study will aid in the diagnosis, management, and treatment choices for patients with LAD-III and ARO.
Collapse
Affiliation(s)
- Tejasvi Dudiki
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniel W Nascimento
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lauren S Childs
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Swetha Kareti
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Charlie Androjna
- Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
10
|
Mead TJ, Martin DR, Wang LW, Cain SA, Gulec C, Cahill E, Mauch J, Reinhardt D, Lo C, Baldock C, Apte SS. Proteolysis of fibrillin-2 microfibrils is essential for normal skeletal development. eLife 2022; 11:71142. [PMID: 35503090 PMCID: PMC9064305 DOI: 10.7554/elife.71142] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 04/13/2022] [Indexed: 01/08/2023] Open
Abstract
The embryonic extracellular matrix (ECM) undergoes transition to mature ECM as development progresses, yet few mechanisms ensuring ECM proteostasis during this period are known. Fibrillin microfibrils are macromolecular ECM complexes serving structural and regulatory roles. In mice, Fbn1 and Fbn2, encoding the major microfibrillar components, are strongly expressed during embryogenesis, but fibrillin-1 is the major component observed in adult tissue microfibrils. Here, analysis of Adamts6 and Adamts10 mutant mouse embryos, lacking these homologous secreted metalloproteases individually and in combination, along with in vitro analysis of microfibrils, measurement of ADAMTS6-fibrillin affinities and N-terminomics discovery of ADAMTS6-cleaved sites, identifies a proteostatic mechanism contributing to postnatal fibrillin-2 reduction and fibrillin-1 dominance. The lack of ADAMTS6, alone and in combination with ADAMTS10 led to excess fibrillin-2 in perichondrium, with impaired skeletal development defined by a drastic reduction of aggrecan and cartilage link protein, impaired BMP signaling in cartilage, and increased GDF5 sequestration in fibrillin-2-rich tissue. Although ADAMTS6 cleaves fibrillin-1 and fibrillin-2 as well as fibronectin, which provides the initial scaffold for microfibril assembly, primacy of the protease-substrate relationship between ADAMTS6 and fibrillin-2 was unequivocally established by reversal of the defects in Adamts6-/- embryos by genetic reduction of Fbn2, but not Fbn1.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering and Musculoskeletal Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Daniel R Martin
- Department of Biomedical Engineering and Musculoskeletal Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Lauren W Wang
- Department of Biomedical Engineering and Musculoskeletal Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Stuart A Cain
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Cagri Gulec
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Elisabeth Cahill
- Department of Biomedical Engineering and Musculoskeletal Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Joseph Mauch
- Department of Biomedical Engineering and Musculoskeletal Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Dieter Reinhardt
- Faculty of Medicine and Health Sciences and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Cecilia Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Clair Baldock
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Suneel S Apte
- Department of Biomedical Engineering and Musculoskeletal Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| |
Collapse
|
11
|
ADAMTS7 Attenuates House Dust Mite-Induced Airway Inflammation and Th2 Immune Responses. Lung 2022; 200:305-313. [PMID: 35503474 PMCID: PMC9205806 DOI: 10.1007/s00408-022-00538-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/15/2022] [Indexed: 10/30/2022]
Abstract
PURPOSE ADAMTS7 is a secreted metalloproteinase enzyme and proteoglycan associated with the early progression of coronary artery disease. However, there is limited information regarding the role of ADAMTS7 in lung adaptive immunity and inflammation. Thus, we sought to assess whether ADAMTS7 expression in the lung modulates house dust mite (HDM)-induced airway inflammation and Th2 immune response. METHODS The role of ADAMTS7 in HDM-induced airway disease was assessed in ADAMTS7-deficient (ADAMTS7-/-) mice and compared with the wild-type control mice by flow cytometry, ELISA, and histopathology. Furthermore, the antigen priming capability of dendritic cells (DC) was determined ex vivo by employing coculture with CD4+ OT-II cells. RESULTS ADAMTS7-/- mice develop an augmented eosinophilic airway inflammation, mucous cell metaplasia, and increased Th2 immune response to inhaled HDM. In addition, allergen uptake by lung DC and migration to draining mediastinal lymph node were significantly increased in ADAMTS7-/- mice, which shows an enhanced capacity to mount allergen-specific T-cell proliferation and effector Th2 cytokine productions. We propose that the mechanism by which ADAMTS7 negatively regulates DC function involves attenuated antigen uptake and presentation capabilities, which reduces allergic sensitization and Th2 immune responses in the lung. CONCLUSION In aggregate, we provide compelling evidence that ADAMTS7 plays a pivotal role in allergic airway disease and Th2 immunity and would be an attractive target for asthma.
Collapse
|
12
|
MacDonald BT, Keshishian H, Mundorff CC, Arduini A, Lai D, Bendinelli K, Popp NR, Bhandary B, Clauser KR, Specht H, Elowe NH, Laprise D, Xing Y, Kaushik VK, Carr SA, Ellinor PT. TAILS Identifies Candidate Substrates and Biomarkers of ADAMTS7, a Therapeutic Protease Target in Coronary Artery Disease. Mol Cell Proteomics 2022; 21:100223. [PMID: 35283288 PMCID: PMC9035411 DOI: 10.1016/j.mcpro.2022.100223] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/05/2022] [Accepted: 03/02/2022] [Indexed: 12/22/2022] Open
Abstract
Loss-of-function mutations in the secreted enzyme ADAMTS7 (a disintegrin and metalloproteinase with thrombospondin motifs 7) are associated with protection for coronary artery disease. ADAMTS7 catalytic inhibition has been proposed as a therapeutic strategy for treating coronary artery disease; however, the lack of an endogenous substrate has hindered the development of activity-based biomarkers. To identify ADAMTS7 extracellular substrates and their cleavage sites relevant to vascular disease, we used TAILS (terminal amine isotopic labeling of substrates), a method for identifying protease-generated neo-N termini. We compared the secreted proteome of vascular smooth muscle and endothelial cells expressing either full-length mouse ADAMTS7 WT, catalytic mutant ADAMTS7 E373Q, or a control luciferase adenovirus. Significantly enriched N-terminal cleavage sites in ADAMTS7 WT samples were compared to the negative control conditions and filtered for stringency, resulting in catalogs of high confidence candidate ADAMTS7 cleavage sites from our three independent TAILS experiments. Within the overlap of these discovery sets, we identified 24 unique cleavage sites from 16 protein substrates, including cleavage sites in EFEMP1 (EGF-containing fibulin-like extracellular matrix protein 1/Fibulin-3). The ADAMTS7 TAILS preference for EFEMP1 cleavage at the amino acids 123.124 over the adjacent 124.125 site was validated using both endogenous EFEMP1 and purified EFEMP1 in a binary in vitro cleavage assay. Collectively, our TAILS discovery experiments have uncovered hundreds of potential substrates and cleavage sites to explore disease-related biological substrates and facilitate activity-based ADAMTS7 biomarker development.
Collapse
Affiliation(s)
- Bryan T MacDonald
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| | - Hasmik Keshishian
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Charles C Mundorff
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alessandro Arduini
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Daniel Lai
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kayla Bendinelli
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nicholas R Popp
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Bidur Bhandary
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Karl R Clauser
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Harrison Specht
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nadine H Elowe
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dylan Laprise
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yi Xing
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Virendar K Kaushik
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Steven A Carr
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
El Masri R, Seffouh A, Roelants C, Seffouh I, Gout E, Pérard J, Dalonneau F, Nishitsuji K, Noborn F, Nikpour M, Larson G, Crétinon Y, Friedel-Arboleas M, Uchimura K, Daniel R, Lortat-Jacob H, Filhol O, Vivès RR. Extracellular endosulfatase Sulf-2 harbors a chondroitin/dermatan sulfate chain that modulates its enzyme activity. Cell Rep 2022; 38:110516. [PMID: 35294879 DOI: 10.1016/j.celrep.2022.110516] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 10/07/2021] [Accepted: 02/17/2022] [Indexed: 11/18/2022] Open
Abstract
Sulfs represent a class of unconventional sulfatases which provide an original post-synthetic regulatory mechanism for heparan sulfate polysaccharides and are involved in multiple physiopathological processes, including cancer. However, Sulfs remain poorly characterized enzymes, with major discrepancies regarding their in vivo functions. Here we show that human Sulf-2 (HSulf-2) harbors a chondroitin/dermatan sulfate glycosaminoglycan (GAG) chain, attached to the enzyme substrate-binding domain. We demonstrate that this GAG chain affects enzyme/substrate recognition and tunes HSulf-2 activity in vitro and in vivo. In addition, we show that mammalian hyaluronidase acts as a promoter of HSulf-2 activity by digesting its GAG chain. In conclusion, our results highlight HSulf-2 as a proteoglycan-related enzyme and its GAG chain as a critical non-catalytic modulator of the enzyme activity. These findings contribute to clarifying the conflicting data on the activities of the Sulfs.
Collapse
Affiliation(s)
- Rana El Masri
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Amal Seffouh
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Caroline Roelants
- Université Grenoble Alpes, INSERM, CEA, IRIG-Biosanté, UMR 1292, 38000 Grenoble, France
| | - Ilham Seffouh
- Université Paris-Saclay, Université Evry, CNRS, LAMBE, 91025 Evry-Courcouronnes, France
| | - Evelyne Gout
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Julien Pérard
- Université Grenoble Alpes, CNRS, IRIG - DIESE - CBM, CEA-Grenoble, 38000 Grenoble, France
| | | | - Kazuchika Nishitsuji
- Department of Biochemistry, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Fredrik Noborn
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mahnaz Nikpour
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yoann Crétinon
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | | | - Kenji Uchimura
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000 Lille, France
| | - Régis Daniel
- Université Paris-Saclay, Université Evry, CNRS, LAMBE, 91025 Evry-Courcouronnes, France
| | | | - Odile Filhol
- Université Grenoble Alpes, INSERM, CEA, IRIG-Biosanté, UMR 1292, 38000 Grenoble, France.
| | - Romain R Vivès
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France.
| |
Collapse
|
14
|
Brachtl G, Poupardin R, Hochmann S, Raninger A, Jürchott K, Streitz M, Schlickeiser S, Oeller M, Wolf M, Schallmoser K, Volk HD, Geissler S, Strunk D. Batch Effects during Human Bone Marrow Stromal Cell Propagation Prevail Donor Variation and Culture Duration: Impact on Genotype, Phenotype and Function. Cells 2022; 11:946. [PMID: 35326396 PMCID: PMC8946746 DOI: 10.3390/cells11060946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Donor variation is a prominent critical issue limiting the applicability of cell-based therapies. We hypothesized that batch effects during propagation of bone marrow stromal cells (BMSCs) in human platelet lysate (hPL), replacing fetal bovine serum (FBS), can affect phenotypic and functional variability. We therefore investigated the impact of donor variation, hPL- vs. FBS-driven propagation and exhaustive proliferation, on BMSC epigenome, transcriptome, phenotype, coagulation risk and osteochondral regenerative function. Notably, propagation in hPL significantly increased BMSC proliferation, created significantly different gene expression trajectories and distinct surface marker signatures, already after just one passage. We confirmed significantly declining proliferative potential in FBS-expanded BMSC after proliferative challenge. Flow cytometry verified the canonical fibroblastic phenotype in culture-expanded BMSCs. We observed limited effects on DNA methylation, preferentially in FBS-driven cultures, irrespective of culture duration. The clotting risk increased over culture time. Moreover, expansion in xenogenic serum resulted in significant loss of function during 3D cartilage disk formation and significantly increased clotting risk. Superior chondrogenic function under hPL-conditions was maintained over culture. The platelet blood group and isoagglutinins had minor impact on BMSC function. These data demonstrate pronounced batch effects on BMSC transcriptome, phenotype and function due to serum factors, partly outcompeting donor variation after just one culture passage.
Collapse
Affiliation(s)
- Gabriele Brachtl
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Rodolphe Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Sarah Hochmann
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Anna Raninger
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Karsten Jürchott
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
| | - Mathias Streitz
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Insel Riems, 17493 Greifswald, Germany
| | - Stephan Schlickeiser
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
| | - Michaela Oeller
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Martin Wolf
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Katharina Schallmoser
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Hans-Dieter Volk
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
- Berlin Center for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Sven Geissler
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
- Berlin Center for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Dirk Strunk
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| |
Collapse
|
15
|
Abstract
A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) are major mediators in extracellular matrix (ECM) turnover and have gained increasing interest over the last years as major players in ECM remodeling during tissue homeostasis and the development of diseases. Although, ADAMTSs are recognized in playing important roles during tissue remodeling, and loss of function in various member of the ADAMTS family could be associated with the development of numerous diseases, limited knowledge is available about their specific substrates and mechanism of action. In this chapter, we will review current knowledge about ADAMTSs and their use as disease biomarkers.
Collapse
Affiliation(s)
- Rahel Schnellmann
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
16
|
Fang F, Sup M, Luzzi A, Ferrer X, Thomopoulos S. Hedgehog signaling underlying tendon and enthesis development and pathology. Matrix Biol 2022; 105:87-103. [PMID: 34954379 PMCID: PMC8821161 DOI: 10.1016/j.matbio.2021.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 02/08/2023]
Abstract
Hedgehog (Hh) signaling has been widely acknowledged to play essential roles in many developmental processes, including endochondral ossification and growth plate maintenance. Furthermore, a rising number of studies have shown that Hh signaling is necessary for tendon enthesis development. Specifically, the well-tuned regulation of Hh signaling during development drives the formation of a mineral gradient across the tendon enthesis fibrocartilage. However, aberrant Hh signaling can also lead to pathologic heterotopic ossification in tendon or osteophyte formation at the enthesis. Therefore, the therapeutic potential of Hh signaling modulation for treating tendon and enthesis diseases remains uncertain. For example, increased Hh signaling may enhance tendon-to-bone healing by promoting the formation of mineralized fibrocartilage at the healing interface, but pathologic heterotopic ossification may also be triggered in the adjacent tendon. Further work is needed to elucidate the distinct functions of Hh signaling in the tendon and enthesis to support the development of therapies that target the pathway.
Collapse
Affiliation(s)
- Fei Fang
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States
| | - McKenzie Sup
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Andrew Luzzi
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States
| | - Xavier Ferrer
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, Black Building, Room 1408, 650W 168 ST, New York, NY 10032-3702, United States; Department of Biomedical Engineering, Columbia University, New York, NY, United States.
| |
Collapse
|
17
|
Mienaltowski MJ, Gonzales NL, Beall JM, Pechanec MY. Basic Structure, Physiology, and Biochemistry of Connective Tissues and Extracellular Matrix Collagens. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:5-43. [PMID: 34807414 DOI: 10.1007/978-3-030-80614-9_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The physiology of connective tissues like tendons and ligaments is highly dependent upon the collagens and other such extracellular matrix molecules hierarchically organized within the tissues. By dry weight, connective tissues are mostly composed of fibrillar collagens. However, several other forms of collagens play essential roles in the regulation of fibrillar collagen organization and assembly, in the establishment of basement membrane networks that provide support for vasculature for connective tissues, and in the formation of extensive filamentous networks that allow for cell-extracellular matrix interactions as well as maintain connective tissue integrity. The structures and functions of these collagens are discussed in this chapter. Furthermore, collagen synthesis is a multi-step process that includes gene transcription, translation, post-translational modifications within the cell, triple helix formation, extracellular secretion, extracellular modifications, and then fibril assembly, fibril modifications, and fiber formation. Each step of collagen synthesis and fibril assembly is highly dependent upon the biochemical structure of the collagen molecules created and how they are modified in the cases of development and maturation. Likewise, when the biochemical structures of collagens or are compromised or these molecules are deficient in the tissues - in developmental diseases, degenerative conditions, or injuries - then the ultimate form and function of the connective tissues are impaired. In this chapter, we also review how biochemistry plays a role in each of the processes involved in collagen synthesis and assembly, and we describe differences seen by anatomical location and region within tendons. Moreover, we discuss how the structures of the molecules, fibrils, and fibers contribute to connective tissue physiology in health, and in pathology with injury and repair.
Collapse
Affiliation(s)
| | - Nicole L Gonzales
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - Jessica M Beall
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - Monica Y Pechanec
- Department of Animal Science, University of California Davis, Davis, CA, USA
| |
Collapse
|
18
|
McMahon M, Ye S, Pedrina J, Dlugolenski D, Stambas J. Extracellular Matrix Enzymes and Immune Cell Biology. Front Mol Biosci 2021; 8:703868. [PMID: 34527702 PMCID: PMC8436118 DOI: 10.3389/fmolb.2021.703868] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022] Open
Abstract
Remodelling of the extracellular matrix (ECM) by ECM metalloproteinases is increasingly being associated with regulation of immune cell function. ECM metalloproteinases, including Matrix Metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs) and ADAMs with Thombospondin-1 motifs (ADAMTS) play a vital role in pathogen defence and have been shown to influence migration of immune cells. This review provides a current summary of the role of ECM enzymes in immune cell migration and function and discusses opportunities and limitations for development of diagnostic and therapeutic strategies targeting metalloproteinase expression and activity in the context of infectious disease.
Collapse
Affiliation(s)
- Meagan McMahon
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Siying Ye
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Jess Pedrina
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Daniel Dlugolenski
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - John Stambas
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
19
|
Rose KWJ, Taye N, Karoulias SZ, Hubmacher D. Regulation of ADAMTS Proteases. Front Mol Biosci 2021; 8:701959. [PMID: 34268335 PMCID: PMC8275829 DOI: 10.3389/fmolb.2021.701959] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
A disintegrin and metalloprotease with thrombospondin type I motifs (ADAMTS) proteases are secreted metalloproteinases that play key roles in the formation, homeostasis and remodeling of the extracellular matrix (ECM). The substrate spectrum of ADAMTS proteases can range from individual ECM proteins to entire families of ECM proteins, such as the hyalectans. ADAMTS-mediated substrate cleavage is required for the formation, remodeling and physiological adaptation of the ECM to the needs of individual tissues and organ systems. However, ADAMTS proteases can also be involved in the destruction of tissues, resulting in pathologies such as arthritis. Specifically, ADAMTS4 and ADAMTS5 contribute to irreparable cartilage erosion by degrading aggrecan, which is a major constituent of cartilage. Arthritic joint damage is a major contributor to musculoskeletal morbidity and the most frequent clinical indication for total joint arthroplasty. Due to the high sequence homology of ADAMTS proteases in their catalytically active site, it remains a formidable challenge to design ADAMTS isotype-specific inhibitors that selectively inhibit ADAMTS proteases responsible for tissue destruction without affecting the beneficial functions of other ADAMTS proteases. In vivo, proteolytic activity of ADAMTS proteases is regulated on the transcriptional and posttranslational level. Here, we review the current knowledge of mechanisms that regulate ADAMTS protease activity in tissues including factors that induce ADAMTS gene expression, consequences of posttranslational modifications such as furin processing, the role of endogenous inhibitors and pharmacological approaches to limit ADAMTS protease activity in tissues, which almost exclusively focus on inhibiting the aggrecanase activity of ADAMTS4 and ADAMTS5.
Collapse
Affiliation(s)
| | | | | | - Dirk Hubmacher
- Orthopaedic Research Laboratories, Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Mizoguchi T, MacDonald BT, Bhandary B, Popp NR, Laprise D, Arduini A, Lai D, Zhu QM, Xing Y, Kaushik VK, Kathiresan S, Ellinor PT. Coronary Disease Association With ADAMTS7 Is Due to Protease Activity. Circ Res 2021; 129:458-470. [PMID: 34176299 DOI: 10.1161/circresaha.121.319163] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Taiji Mizoguchi
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA.,Now with Verve Therapeutics, Cambridge, MA, USA (T.M., S.K.)
| | - Bryan T MacDonald
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Bidur Bhandary
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Nicholas R Popp
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Dylan Laprise
- Center for the Development of Therapeutics (D.L., Y.X., V.K.K.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Alessandro Arduini
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Daniel Lai
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Qiuyu Martin Zhu
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA.,Center for Genomic Medicine (Q.M.Z., S.K.), Massachusetts General Hospital, Boston
| | - Yi Xing
- Center for the Development of Therapeutics (D.L., Y.X., V.K.K.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Virendar K Kaushik
- Center for the Development of Therapeutics (D.L., Y.X., V.K.K.), Broad Institute of MIT and Harvard, Cambridge, MA
| | - Sekar Kathiresan
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA.,Now with Verve Therapeutics, Cambridge, MA, USA (T.M., S.K.).,Center for Genomic Medicine (Q.M.Z., S.K.), Massachusetts General Hospital, Boston
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative (T.M., B.T.M., B.B., N.R.P., A.A., D.L., Q.M.Z., S.K., P.T.E.), Broad Institute of MIT and Harvard, Cambridge, MA.,Cardiovascular Research Center (P.T.E.), Massachusetts General Hospital, Boston
| |
Collapse
|
21
|
Mohamedi Y, Fontanil T, Cal S, Cobo T, Obaya ÁJ. ADAMTS-12: Functions and Challenges for a Complex Metalloprotease. Front Mol Biosci 2021; 8:686763. [PMID: 33996918 PMCID: PMC8119882 DOI: 10.3389/fmolb.2021.686763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Nineteen members of the ADAMTS family of secreted zinc metalloproteinases are present in the human degradome. A wide range of different functions are being attributed to these enzymes and the number of their known substrates is considerably increasing in recent years. ADAMTSs can participate in processes such as fertility, inflammation, arthritis, neuronal and behavioral disorders, as well as cancer. Since its first annotation in 2001, ADAMTS-12 has been described to participate in different processes displayed by members of this family of proteinases. In this sense, ADAMTS-12 performs essential roles in modulation and recovery from inflammatory processes such as colitis, endotoxic sepsis and pancreatitis. ADAMTS-12 has also been involved in cancer development acting either as a tumor suppressor or as a pro-tumoral agent. Furthermore, participation of ADAMTS-12 in arthritis or in neuronal disorders has also been suggested through degradation of components of the extracellular matrix. In addition, ADAMTS-12 proteinase activity can also be modified by interaction with other proteins and thus, can be an alternative way of modulating ADAMTS-12 functions. In this review we revised the most relevant findings about ADAMTS-12 function on the 20th anniversary of its identification.
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Departamento de Investigación, Instituto Ordóñez, Oviedo, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, Oviedo, Spain.,Instituto Asturiano de Odontología, Oviedo, Spain
| | - Álvaro J Obaya
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain.,Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
22
|
Bobzin L, Roberts RR, Chen HJ, Crump JG, Merrill AE. Development and maintenance of tendons and ligaments. Development 2021; 148:239823. [PMID: 33913478 DOI: 10.1242/dev.186916] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tendons and ligaments are fibrous connective tissues vital to the transmission of force and stabilization of the musculoskeletal system. Arising in precise regions of the embryo, tendons and ligaments share many properties and little is known about the molecular differences that differentiate them. Recent studies have revealed heterogeneity and plasticity within tendon and ligament cells, raising questions regarding the developmental mechanisms regulating tendon and ligament identity. Here, we discuss recent findings that contribute to our understanding of the mechanisms that establish and maintain tendon progenitors and their differentiated progeny in the head, trunk and limb. We also review the extent to which these findings are specific to certain anatomical regions and model organisms, and indicate which findings similarly apply to ligaments. Finally, we address current research regarding the cellular lineages that contribute to tendon and ligament repair, and to what extent their regulation is conserved within tendon and ligament development.
Collapse
Affiliation(s)
- Lauren Bobzin
- Division of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ryan R Roberts
- Division of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hung-Jhen Chen
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amy E Merrill
- Division of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
23
|
Satz-Jacobowitz B, Hubmacher D. The quest for substrates and binding partners: A critical barrier for understanding the role of ADAMTS proteases in musculoskeletal development and disease. Dev Dyn 2020; 250:8-26. [PMID: 32875613 DOI: 10.1002/dvdy.248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/16/2022] Open
Abstract
Secreted ADAMTS metalloproteases are involved in the sculpting, remodeling, and erosion of connective tissues throughout the body, including in the musculoskeletal system. ADAMTS proteases contribute to musculoskeletal development, pathological tissue destruction, and are mutated in congenital musculoskeletal disorders. Examples include versican cleavage by ADAMTS9 which is required for interdigital web regression during limb development, ADAMTS5-mediated aggrecan degradation in osteoarthritis resulting in joint erosion, and mutations in ADAMTS10 or ADAMTS17 that cause Weill-Marchesani syndrome, a short stature syndrome with bone, joint, muscle, cardiac, and eye involvement. Since the function of ADAMTS proteases and proteases in general is primarily defined by the molecular consequences of proteolysis of their respective substrates, it is paramount to identify all physiological substrates for each individual ADAMTS protease. Here, we review the current knowledge of ADAMTS proteases and their involvement in musculoskeletal development and disease, focusing on some of their known physiological substrates and the consequences of substrate cleavage. We further emphasize the critical need for the identification and validation of novel ADAMTS substrates and binding partners by describing the principles of mass spectrometry-based approaches and by emphasizing strategies that need to be considered for validating the physiological relevance for ADAMTS-mediated proteolysis of novel putative substrates.
Collapse
Affiliation(s)
- Brandon Satz-Jacobowitz
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dirk Hubmacher
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
24
|
Wang C, Chen Y, Xiang H, Wu X, Tang Q, Ma X, Zhang L. ADAMTS7 degrades Comp to fuel BMP2-dependent osteogenic differentiation and ameliorate oncogenic potential in osteosarcomas. FEBS Open Bio 2020; 10:1856-1867. [PMID: 32692461 PMCID: PMC7459396 DOI: 10.1002/2211-5463.12939] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/23/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents, with a high metastatic potential. Despite dramatic changes in OS treatments over the past decades, their efficiency still remains limited, with severe complications and adverse side effects. Key mechanisms underlining tumorigenesis, metastasis and chemotherapy resistance are currently lacking, in turn hindering any progress with respect to developing effective and safe therapeutic strategies against OS. Recently, ADAMTS7, a member of the disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, was shown to be involved in osteogenic differentiation‐related pathological processes. ADAMTS7 promotes vascular calcification via disturbing the balance between osteogenic bone morphogenetic protein (BMP)2 (regulating osteogenic differentiation and bone formation during development) and its natural inhibitor cartilage oligomeric matrix protein (Comp). Hence, in the present study, we aimed to investigate the role of ADAMTS7 in the pathological process of OS. We first revealed that ADAMTS7 was decreased in OS tissues. Lower expression of ADAMTS7 was correlated with poor histological differentiation and an advanced clinical stage of OS. Through loss‐ and gain‐function analysis, we further revealed that ADAMTS7 attenuated cell proliferation, migration and invasion, at the same time as promoting the expression of osteogenic differentiation markers in two OS cell lines: MG63 and SAOS2. Moreover, Comp was responsible for the effects of ADAMTS7 on OS pathogenesis by reinforcing cell osteogenic differentiation mediated by BMP2 in vitro. In conclusion, ADAMTS7‐mediated degradation of Comp may provide a potential therapeutic target for the treatment of OS.
Collapse
Affiliation(s)
- Chao Wang
- Department of Spine SurgeryThe Affiliated Hospital of Qingdao UniversityChina
| | - Yunqing Chen
- Department of PathologyThe Affiliated Hospital of Qingdao UniversityChina
| | - Hongfei Xiang
- Department of Spine SurgeryThe Affiliated Hospital of Qingdao UniversityChina
| | - Xiaolin Wu
- Department of Spine SurgeryThe Affiliated Hospital of Qingdao UniversityChina
| | - Qian Tang
- Department of Medical GeneticsThe Affiliated Hospital of Qingdao UniversityChina
- Medicine Research CenterThe Affiliated Hospital of Qingdao UniversityChina
| | - Xuexiao Ma
- Department of Spine SurgeryThe Affiliated Hospital of Qingdao UniversityChina
| | - Lu Zhang
- Department of Medical GeneticsThe Affiliated Hospital of Qingdao UniversityChina
- Medicine Research CenterThe Affiliated Hospital of Qingdao UniversityChina
| |
Collapse
|
25
|
From hepatitis C virus immunoproteomics to rheumatology via cross-reactivity in one table. Curr Opin Rheumatol 2020; 31:488-492. [PMID: 31356379 DOI: 10.1097/bor.0000000000000606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW To give an overview of molecular and immunologic data that link hepatitis C virus (HCV) infection to rheumatic diseases in the human host. RECENT FINDINGS A high level of peptide sharing exists between immunopositive HCV epitopes and human proteins that, when altered, associate with rheumatic manifestations. SUMMARY The findings suggest the involvement of HCV infection in the induction of most rheumatic diseases via a mechanism of autoimmune cross-reactivity.
Collapse
|
26
|
Karoulias SZ, Taye N, Stanley S, Hubmacher D. The ADAMTS/Fibrillin Connection: Insights into the Biological Functions of ADAMTS10 and ADAMTS17 and Their Respective Sister Proteases. Biomolecules 2020; 10:biom10040596. [PMID: 32290605 PMCID: PMC7226509 DOI: 10.3390/biom10040596] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/28/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022] Open
Abstract
Secreted adisintegrin-like and metalloprotease with thrombospondin type 1 motif (ADAMTS) proteases play crucial roles in tissue development and homeostasis. The biological and pathological functions of ADAMTS proteases are determined broadly by their respective substrates and their interactions with proteins in the pericellular and extracellular matrix. For some ADAMTS proteases, substrates have been identified and substrate cleavage has been implicated in tissue development and in disease. For other ADAMTS proteases, substrates were discovered in vitro, but the role of these proteases and the consequences of substrate cleavage in vivo remains to be established. Mutations in ADAMTS10 and ADAMTS17 cause Weill–Marchesani syndrome (WMS), a congenital syndromic disorder that affects the musculoskeletal system (short stature, pseudomuscular build, tight skin), the eyes (lens dislocation), and the heart (heart valve abnormalities). WMS can also be caused by mutations in fibrillin-1 (FBN1), which suggests that ADAMTS10 and ADAMTS17 cooperate with fibrillin-1 in a common biological pathway during tissue development and homeostasis. Here, we compare and contrast the biochemical properties of ADAMTS10 and ADAMTS17 and we summarize recent findings indicating potential biological functions in connection with fibrillin microfibrils. We also compare ADAMTS10 and ADAMTS17 with their respective sister proteases, ADAMTS6 and ADAMTS19; both were recently linked to human disorders distinct from WMS. Finally, we propose a model for the interactions and roles of these four ADAMTS proteases in the extracellular matrix.
Collapse
|
27
|
Abstract
Understanding proteolytic remodeling of extracellular matrix involves the generation of global or conditional knockout mice by homologous recombination in embryonic stem cells or their manipulation through new advanced technologies such as CRISPR-Cas9. These models provide opportunities to understand the roles of ADAMTS genes in skeletogenesis. Whole-mount skeletal preparations are necessary for assessment of the skeletal phenotype. They allow for facile visualization of skeletal patterning, size and shape of skeletal elements, and skeletal structure. This protocol describes the staining of the murine skeleton using Alcian blue to identify cartilage and alizarin red to identify bone.
Collapse
|
28
|
Abstract
RNA in situ hybridization has an important place in matrix biology, as the only method that allows for in situ discrimination of precise spatial and temporal patterns of gene expression. Whereas immunohistochemistry shows where a matrix protein localizes, ISH identifies the cell of origin. Thus, these methods provide complementary information for insights on the life cycle of matrix molecules, including ADAMTS proteases. This protocol encompasses the staining of tissue sections to reveal expression of the gene of interest.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Suneel S Apte
- Department of Biomedical Engineering-ND20, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
29
|
Taye N, Karoulias SZ, Hubmacher D. The "other" 15-40%: The Role of Non-Collagenous Extracellular Matrix Proteins and Minor Collagens in Tendon. J Orthop Res 2020; 38:23-35. [PMID: 31410892 PMCID: PMC6917864 DOI: 10.1002/jor.24440] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/02/2019] [Indexed: 02/04/2023]
Abstract
Extracellular matrix (ECM) determines the physiological function of all tissues, including musculoskeletal tissues. In tendon, ECM provides overall tissue architecture, which is tailored to match the biomechanical requirements of their physiological function, that is, force transmission from muscle to bone. Tendon ECM also constitutes the microenvironment that allows tendon-resident cells to maintain their phenotype and that transmits biomechanical forces from the macro-level to the micro-level. The structure and function of adult tendons is largely determined by the hierarchical organization of collagen type I fibrils. However, non-collagenous ECM proteins such as small leucine-rich proteoglycans (SLRPs), ADAMTS proteases, and cross-linking enzymes play critical roles in collagen fibrillogenesis and guide the hierarchical bundling of collagen fibrils into tendon fascicles. Other non-collagenous ECM proteins such as the less abundant collagens, fibrillins, or elastin, contribute to tendon formation or determine some of their biomechanical properties. The interfascicular matrix or endotenon and the outer layer of tendons, the epi- and paratenon, includes collagens and non-collagenous ECM proteins, but their function is less well understood. The ECM proteins in the epi- and paratenon may provide the appropriate microenvironment to maintain the identity of distinct tendon cell populations that are thought to play a role during repair processes after injury. The aim of this review is to provide an overview of the role of non-collagenous ECM proteins and less abundant collagens in tendon development and homeostasis. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:23-35, 2020.
Collapse
Affiliation(s)
- Nandaraj Taye
- Leni & Peter W. May Department of Orthopaedics, Orthopaedic Research LaboratoriesIcahn School of Medicine at Mt. SinaiNew York New York 10029
| | - Stylianos Z. Karoulias
- Leni & Peter W. May Department of Orthopaedics, Orthopaedic Research LaboratoriesIcahn School of Medicine at Mt. SinaiNew York New York 10029
| | - Dirk Hubmacher
- Leni & Peter W. May Department of Orthopaedics, Orthopaedic Research LaboratoriesIcahn School of Medicine at Mt. SinaiNew York New York 10029
| |
Collapse
|
30
|
Abstract
ADAMTS7 is a secreted protease that is predominantly expressed in tissues of the cardiovascular system and tendon. Although recent evidence suggests that it may play a role in the etiology of coronary artery disease, its physiological function and substrates are unknown. The enzyme undergoes extensive posttranslational modifications, including chondroitin sulfate attachment, N and O-linked glycosylation, and a two-step activation process. For the benefit of scientists who study the function of ADAMTS7 and its role in disease, this chapter provides an introduction to the chemical and functional properties of the various ADAMTS7 domains, as well as a protocol for the recombinant expression and purification of ADAMTS7.
Collapse
Affiliation(s)
- Rens de Groot
- Center for Hematology, Imperial College, London, UK.
| |
Collapse
|
31
|
Abstract
The ADAMTS superfamily comprises secreted metalloproteases (ADAMTS proteases) as well as structurally related secreted glycoproteins that lack catalytic activity (ADAMTS-like proteins). Members of both families participate in diverse morphogenetic processes during embryonic development, and connective tissue maintenance and hemostasis in the adult. Several ADAMTS proteins are heavily implicated in genetic and acquired human and animal disorders. Despite these indicators of a profound biological and medical importance, detailed knowledge about their molecular structures, substrates, biological pathways, and biochemical mechanisms is significantly limited by unique intrinsic characteristics, which have led to several technical challenges. As a group, they are larger, more heavily modified, and harder to purify than other secreted proteases. In addition, idiosyncratic aspects of individual members are deserving of further investigation but can complicate their analysis. Here, some of the key concepts, challenges, and prospects in ADAMTS research are discussed in the context of the knowledge accumulated over the past two decades. Individual chapters in this volume of Methods in Molecular Biology provide practical solutions for surmounting these challenges. Since the biology of a protease is actually the biology of its substrates, there is considerable emphasis on purification of recombinant ADAMTS proteins, identification of their substrates and assays for their proteolytic activity.
Collapse
|
32
|
Karoulias SZ, Beyens A, Balic Z, Symoens S, Vandersteen A, Rideout AL, Dickinson J, Callewaert B, Hubmacher D. A novel ADAMTS17 variant that causes Weill-Marchesani syndrome 4 alters fibrillin-1 and collagen type I deposition in the extracellular matrix. Matrix Biol 2019; 88:1-18. [PMID: 31726086 DOI: 10.1016/j.matbio.2019.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 12/17/2022]
Abstract
Weill-Marchesani syndrome (WMS) is a rare genetic disorder that affects the musculoskeletal system, the eye, and the cardiovascular system. Individuals with WMS present with short stature, joint contractures, thick skin, microspherophakia, small and dislocated lenses, and cardiac valve anomalies. WMS can be caused by recessive mutations in ADAMTS10 (WMS 1), ADAMTS17 (WMS 4), or LTBP2 (WMS 3), or by dominant mutations in fibrillin-1 (FBN1) (WMS 2); all genes encode secreted extracellular matrix (ECM) proteins. Individuals with WMS 4 due to ADAMTS17 mutations appear to have less severe cardiac involvement and present predominantly with the musculoskeletal and ocular features of WMS. ADAMTS17 is a member of the ADAMTS family of secreted proteases and directly binds to fibrillins. Here we report a novel pathogenic variant in ADAMTS17 that causes WMS 4 in an individual with short stature, brachydactyly, and small, spherical, and dislocated lenses. We provide biochemical and cell biological insights in the pathomechanisms of WMS 4, which also suggest potential biological functions for ADAMTS17. We show that the variant in ADAMTS17 prevents its secretion and we found intracellular accumulation of fibrillin-1 and collagen type I in patient-derived skin fibroblasts. In accordance, transmission electron microscopy revealed elastic fiber abnormalities, decreased collagen fibril diameters, and intracellular collagen accumulation in the dermis of the proband. Together, the data indicate a possible role for ADAMTS17 in the secretion of fibrillin-1 and collagen type I or in their early assembly in the pericellular matrix or the ECM.
Collapse
Affiliation(s)
- Stylianos Z Karoulias
- Orthopaedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Aude Beyens
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Belgium; Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - Zerina Balic
- Orthopaedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Sofie Symoens
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Belgium
| | - Anthony Vandersteen
- Division of Medical Genetics, Department of Pediatrics, Dalhousie University, Halifax, NS, Canada; Maritime Medical Genetics Service, IWK Health Centre, Halifax, NS, Canada
| | - Andrea L Rideout
- Maritime Medical Genetics Service, IWK Health Centre, Halifax, NS, Canada
| | - John Dickinson
- Department of Ophthalmology & Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Belgium.
| | - Dirk Hubmacher
- Orthopaedic Research Laboratories, Leni & Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mt. Sinai, New York, NY, USA.
| |
Collapse
|
33
|
Colige A, Monseur C, Crawley JTB, Santamaria S, de Groot R. Proteomic discovery of substrates of the cardiovascular protease ADAMTS7. J Biol Chem 2019; 294:8037-8045. [PMID: 30926607 PMCID: PMC6527163 DOI: 10.1074/jbc.ra119.007492] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/28/2019] [Indexed: 12/23/2022] Open
Abstract
The protease ADAMTS7 functions in the extracellular matrix (ECM) of the cardiovascular system. However, its physiological substrate specificity and mechanism of regulation remain to be explored. To address this, we conducted an unbiased substrate analysis using terminal amine isotopic labeling of substrates (TAILS). The analysis identified candidate substrates of ADAMTS7 in the human fibroblast secretome, including proteins with a wide range of functions, such as collagenous and noncollagenous extracellular matrix proteins, growth factors, proteases, and cell-surface receptors. It also suggested that autolysis occurs at Glu-729-Val-730 and Glu-732-Ala-733 in the ADAMTS7 Spacer domain, which was corroborated by N-terminal sequencing and Western blotting. Importantly, TAILS also identified proteolysis of the latent TGF-β-binding proteins 3 and 4 (LTBP3/4) at a Glu-Val and Glu-Ala site, respectively. Using purified enzyme and substrate, we confirmed ADAMTS7-catalyzed proteolysis of recombinant LTBP4. Moreover, we identified multiple additional scissile bonds in an N-terminal linker region of LTBP4 that connects fibulin-5/tropoelastin and fibrillin-1-binding regions, which have an important role in elastogenesis. ADAMTS7-mediated cleavage of LTBP4 was efficiently inhibited by the metalloprotease inhibitor TIMP-4, but not by TIMP-1 and less efficiently by TIMP-2 and TIMP-3. As TIMP-4 expression is prevalent in cardiovascular tissues, we propose that TIMP-4 represents the primary endogenous ADAMTS7 inhibitor. In summary, our findings reveal LTBP4 as an ADAMTS7 substrate, whose cleavage may potentially impact elastogenesis in the cardiovascular system. We also identify TIMP-4 as a likely physiological ADAMTS7 inhibitor.
Collapse
Affiliation(s)
- Alain Colige
- Laboratory of Connective Tissue Biology, GIGA, University of Liège, Sart-Tilman, 4000 Liège, Belgium
| | - Christine Monseur
- Laboratory of Connective Tissue Biology, GIGA, University of Liège, Sart-Tilman, 4000 Liège, Belgium
| | - James T B Crawley
- Centre for Haematology, Imperial College London, W12 0NN London, United Kingdom
| | | | - Rens de Groot
- Centre for Haematology, Imperial College London, W12 0NN London, United Kingdom.
| |
Collapse
|
34
|
Design, synthesis and biological evaluation of inhibitors of cathepsin K on dedifferentiated chondrocytes. Bioorg Med Chem 2019; 27:1034-1042. [DOI: 10.1016/j.bmc.2019.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/27/2019] [Accepted: 02/01/2019] [Indexed: 01/18/2023]
|
35
|
Wang LW, Kutz WE, Mead TJ, Beene LC, Singh S, Jenkins MW, Reinhardt DP, Apte SS. Adamts10 inactivation in mice leads to persistence of ocular microfibrils subsequent to reduced fibrillin-2 cleavage. Matrix Biol 2018; 77:117-128. [PMID: 30201140 DOI: 10.1016/j.matbio.2018.09.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 02/02/2023]
Abstract
Mutations in the secreted metalloproteinase ADAMTS10 cause recessive Weill-Marchesani syndrome (WMS), comprising ectopia lentis, short stature, brachydactyly, thick skin and cardiac valve anomalies. Dominant WMS caused by FBN1 mutations is clinically similar and affects fibrillin-1 microfibrils, which are a major component of the ocular zonule. ADAMTS10 was previously shown to enhance fibrillin-1 assembly in vitro. Here, Adamts10 null mice were analyzed to determine the impact of ADAMTS10 deficiency on fibrillin microfibrils in vivo. An intragenic lacZ reporter identified widespread Adamts10 expression in the eye, musculoskeletal tissues, vasculature, skin and lung. Adamts10-/- mice had reduced viability on the C57BL/6 background, and although surviving mice were slightly smaller and had stiff skin, they lacked brachydactyly and cardiovascular defects. Ectopia lentis was not observed in Adamts10-/- mice, similar to Fbn1-/- mice, most likely because the mouse zonule contains fibrillin-2 in addition to fibrillin-1. Unexpectedly, in contrast to wild-type eyes, Adamts10-/- zonule fibers were thicker and immunostained strongly with fibrillin-2 antibodies into adulthood, whereas fibrillin-1 staining was reduced. Furthermore, fibrillin-2 staining of hyaloid vasculature remnants persisted post-natally in Adamts10-/- eyes. ADAMTS10 was found to cleave fibrillin-2, providing an explanation for persistence of fibrillin-2 at these sites. Thus, analysis of Adamts10-/- mice led to identification of fibrillin-2 as a novel ADAMTS10 substrate and defined a proteolytic mechanism for clearance of ocular fibrillin-2 at the end of the juvenile period.
Collapse
Affiliation(s)
- Lauren W Wang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Wendy E Kutz
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy J Mead
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lauren C Beene
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shweta Singh
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Michael W Jenkins
- Department of Pediatrics and Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dieter P Reinhardt
- Department of Anatomy and Cell Biology and Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Suneel S Apte
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
36
|
Mead TJ, Apte SS. ADAMTS proteins in human disorders. Matrix Biol 2018; 71-72:225-239. [PMID: 29885460 DOI: 10.1016/j.matbio.2018.06.002] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023]
Abstract
ADAMTS proteins are a superfamily of 26 secreted molecules comprising two related, but distinct families. ADAMTS proteases are zinc metalloendopeptidases, most of whose substrates are extracellular matrix (ECM) components, whereas ADAMTS-like proteins lack a metalloprotease domain, reside in the ECM and have regulatory roles vis-à-vis ECM assembly and/or ADAMTS activity. Evolutionary conservation and expansion of ADAMTS proteins in mammals is suggestive of crucial embryologic or physiological roles in humans. Indeed, Mendelian disorders or birth defects resulting from naturally occurring ADAMTS2, ADAMTS3, ADAMTS10, ADAMTS13, ADAMTS17, ADAMTS20, ADAMTSL2 and ADAMTSL4 mutations as well as numerous phenotypes identified in genetically engineered mice have revealed ADAMTS participation in major biological pathways. Important roles have been identified in a few acquired conditions. ADAMTS5 is unequivocally implicated in pathogenesis of osteoarthritis via degradation of aggrecan, a major structural proteoglycan in cartilage. ADAMTS7 is strongly associated with coronary artery disease and promotes atherosclerosis. Autoantibodies to ADAMTS13 lead to a platelet coagulopathy, thrombotic thrombocytopenic purpura, which is similar to that resulting from ADAMTS13 mutations. ADAMTS proteins have numerous potential connections to other human disorders that were identified by genome-wide association studies. Here, we review inherited and acquired human disorders in which ADAMTS proteins participate, and discuss progress and prospects in therapeutics.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, United States
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, United States.
| |
Collapse
|