1
|
Renaud L, Waldrep KM, da Silveira WA, Pilewski JM, Feghali-Bostwick CA. First Characterization of the Transcriptome of Lung Fibroblasts of SSc Patients and Healthy Donors of African Ancestry. Int J Mol Sci 2023; 24:3645. [PMID: 36835058 PMCID: PMC9966000 DOI: 10.3390/ijms24043645] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/25/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disorder that results in fibrosis of the skin and visceral organs. SSc-associated pulmonary fibrosis (SSc-PF) is the leading cause of death amongst SSc patients. Racial disparity is noted in SSc as African Americans (AA) have a higher frequency and severity of disease than European Americans (EA). Using RNAseq, we determined differentially expressed genes (DEGs; q < 0.1, log2FC > |0.6|) in primary pulmonary fibroblasts from SSc lungs (SScL) and normal lungs (NL) of AA and EA patients to characterize the unique transcriptomic signatures of AA-NL and AA-SScL fibroblasts using systems-level analysis. We identified 69 DEGs in "AA-NL vs. EA-NL" and 384 DEGs in "AA-SScL vs. EA-SScL" analyses, and a comparison of disease mechanisms revealed that only 7.5% of DEGs were commonly deregulated in AA and EA patients. Surprisingly, we also identified an SSc-like signature in AA-NL fibroblasts. Our data highlight differences in disease mechanisms between AA and EA SScL fibroblasts and suggest that AA-NL fibroblasts are in a "pre-fibrosis" state, poised to respond to potential fibrotic triggers. The DEGs and pathways identified in our study provide a wealth of novel targets to better understand disease mechanisms leading to racial disparity in SSc-PF and develop more effective and personalized therapies.
Collapse
Affiliation(s)
- Ludivine Renaud
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristy M. Waldrep
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Willian A. da Silveira
- Department of Biological Sciences, School of Life Sciences and Education, Staffordshire University, Stoke-on-Trent ST4 2DF, UK
| | - Joseph M. Pilewski
- Department of Medicine, Pulmonary, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Carol A. Feghali-Bostwick
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Plaas AHK, Moran MM, Sandy JD, Hascall VC. Aggrecan and Hyaluronan: The Infamous Cartilage Polyelectrolytes - Then and Now. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:3-29. [PMID: 37052843 DOI: 10.1007/978-3-031-25588-5_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Cartilages are unique in the family of connective tissues in that they contain a high concentration of the glycosaminoglycans, chondroitin sulfate and keratan sulfate attached to the core protein of the proteoglycan, aggrecan. Multiple aggrecan molecules are organized in the extracellular matrix via a domain-specific molecular interaction with hyaluronan and a link protein, and these high molecular weight aggregates are immobilized within the collagen and glycoprotein network. The high negative charge density of glycosaminoglycans provides hydrophilicity, high osmotic swelling pressure and conformational flexibility, which together function to absorb fluctuations in biomechanical stresses on cartilage during movement of an articular joint. We have summarized information on the history and current knowledge obtained by biochemical and genetic approaches, on cell-mediated regulation of aggrecan metabolism and its role in skeletal development, growth as well as during the development of joint disease. In addition, we describe the pathways for hyaluronan metabolism, with particular focus on the role as a "metabolic rheostat" during chondrocyte responses in cartilage remodeling in growth and disease.Future advances in effective therapeutic targeting of cartilage loss during osteoarthritic diseases of the joint as an organ as well as in cartilage tissue engineering would benefit from 'big data' approaches and bioinformatics, to uncover novel feed-forward and feed-back mechanisms for regulating transcription and translation of genes and their integration into cell-specific pathways.
Collapse
Affiliation(s)
- Anna H K Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | - Meghan M Moran
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - John D Sandy
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
3
|
Eremenko E, Ding J, Kwan P, Tredget EE. The Biology of Extracellular Matrix Proteins in Hypertrophic Scarring. Adv Wound Care (New Rochelle) 2022; 11:234-254. [PMID: 33913776 DOI: 10.1089/wound.2020.1257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Significance: Hypertrophic scars (HTS) are a fibroproliferative disorder that occur following deep dermal injury and affect up to 72% of burn patients. These scars result in discomfort, impaired mobility, disruption of normal function and cosmesis, and significant psychological distress. Currently, there are no satisfactory methods to treat or prevent HTS, as the cellular and molecular mechanisms are complex and incompletely understood. This review summarizes the biology of proteins in the dermal extracellular matrix (ECM), which are involved in wound healing and hypertrophic scarring. Recent Advances: New basic research continues toward understanding the diversity of cellular and molecular mechanisms of normal wound healing and hypertrophic scarring. Broadening the understanding of these mechanisms creates insight into novel methods for preventing and treating HTS. Critical Issues: Although there is an abundance of research conducted on collagen in the ECM and its relationship to HTS, there is a significant gap in understanding the role of proteoglycans and their specific isoforms in dermal fibrosis. Future Directions: Exploring the biological roles of ECM proteins and their unique isoforms in HTS, mature scars, and normal skin will further the understanding of abnormal wound healing and create a more robust understanding of what constitutes dermal fibrosis. Research into the biological roles of ECM protein isoforms and their regulation during wound healing warrants a more extensive investigation to identify their distinct biological functions in cellular processes and outcomes.
Collapse
Affiliation(s)
- Elizabeth Eremenko
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Peter Kwan
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Edward E. Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| |
Collapse
|
4
|
Bolia IK, Mertz K, Faye E, Sheppard J, Telang S, Bogdanov J, Hasan LK, Haratian A, Evseenko D, Weber AE, Petrigliano FA. Cross-Communication Between Knee Osteoarthritis and Fibrosis: Molecular Pathways and Key Molecules. Open Access J Sports Med 2022; 13:1-15. [PMID: 35261547 PMCID: PMC8898188 DOI: 10.2147/oajsm.s321139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/18/2022] [Indexed: 01/26/2023] Open
Abstract
Knee fibrosis is characterized by the presence of excessive connective tissue due to dysregulated fibroblast activation following local or systemic tissue damage. Knee fibrosis constitutes a major clinical problem in orthopaedics due to the severe limitation in the knee range of motion that leads to compromised function and patient disability. Knee osteoarthritis is an extremely common orthopedic condition that is associated with patient disability and major costs to the health-care systems worldwide. Although knee fibrosis and osteoarthritis (OA) have traditionally been perceived as two separate pathologic entities, recent research has shown common ground between the pathophysiologic processes that lead to the development of these two conditions. The purpose of this review was to identify the pathophysiologic pathways as well as key molecules that are implicated in the development of both knee OA and knee fibrosis in order to understand the relationship between the two diagnoses and potentially identify novel therapeutic targets.
Collapse
Affiliation(s)
- Ioanna K Bolia
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA,Correspondence: Ioanna K Bolia, 1520 San Pablo Street Suite 2000, Los Angeles, CA, 90033, USA, Tel +1 9703432813, Fax +1 818-658-5925, Email
| | - Kevin Mertz
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Ethan Faye
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Justin Sheppard
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Sagar Telang
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Jacob Bogdanov
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Laith K Hasan
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Aryan Haratian
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Denis Evseenko
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Alexander E Weber
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Frank A Petrigliano
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| |
Collapse
|
5
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
6
|
Connective Tissue Disorders in Domestic Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:325-335. [PMID: 34807427 DOI: 10.1007/978-3-030-80614-9_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Though soft tissue disorders have been recognized and described to some detail in several types of domestic animals and small mammals for some years, they remain uncommon. Because of their low prevalence, not much progress has been made not only in improved diagnosis but also in our understanding of the biochemical basis and pathogenesis of these diseases in animals. Ehlers-Danlos syndrome (EDS) described in dogs already in 1943 and later in cats has only minor impact on the well-being of the dog as its effects on skin of these animals are rather limited. The involved skin is thin and hyperextensible with easily inflicted injuries resulting in hemorrhagic wounds and atrophic scars. Joint laxity and dislocation common in people are less frequently found in dogs. No systemic complications, such as organ rupture or cardiovascular problems which have devastating consequences in people have been described in cats and dogs. The diagnosis is based on clinical presentation and on light or electron microscopic features of disorganized and fragmented collagen fibrils. Several case of bovine and ovine dermatosparaxis analogous to human Ehlers-Danlos syndrome type VIIC were found to be caused by mutations in the procollagen I N-proteinase (pnPI) or ADAMTS2 gene, though mutations in other sites are likely responsible for other types of dermatosparaxis. Cattle suffering from a form of Marfan syndrome (MFS) were described to have aortic dilatation and aneurysm together with ocular abnormalities and skeletal involvement. As in people, mutations at different sites of bovine FBN1 may be responsible for Marfan phenotype. Hereditary equine regional dermal asthenia (HERDA), or hyperelastosis cutis, has been recognized in several horse breeds as affecting primarily skin, and, occasionally, tendons. A mutation in cyclophilin B, a chaperon involved in proper folding of collagens, has been identified in some cases. Warmblood fragile foal syndrome (WFFS) is another Ehlers-Danlos-like disorder in horses, affecting primarily Warmbloods who present with skin fragility and joint hyperextensibility. Degenerative suspensory ligament desmitis (DSLD) affects primarily tendons and ligaments of certain horse breeds. Data from our laboratory showed excessive accumulation of proteoglycans in organs with high content of connective tissues. We have identified increased presence of bone morphogenetic protein 2 (BMP2) in active foci of DSLD and an abnormal form of decorin in proteoglycan deposits. Our most recent data obtained from next generation sequencing showed disturbances in expression of genes for numerous proteoglycans and collagens.
Collapse
|
7
|
Isolation and Purification of Versican and Analysis of Versican Proteolysis. Methods Mol Biol 2021. [PMID: 34626407 DOI: 10.1007/978-1-0716-1398-6_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Versican is a widely distributed chondroitin sulfate proteoglycan that forms large complexes with the glycosaminoglycan hyaluronan (HA). As a consequence of HA binding to its receptor CD44 and interactions of the versican C-terminal globular (G3) domain with a variety of extracellular matrix proteins, versican is a key component of well-defined networks in pericellular matrix and extracellular matrix. Versican is crucial for several developmental processes in the embryo ranging from cardiac development to digit separation, and there is an increasing interest in its roles in cancer and inflammation. Versican proteolysis by ADAMTS proteases is highly regulated, occurs at specific peptide bonds, and is relevant to several physiological and disease mechanisms. In this chapter, methods are described for the isolation and detection of intact and cleaved versican in tissues using morphologic and biochemical techniques. These, together with the methodologies for purification and analysis of recombinant versican and an N-terminal versican fragment named versikine that are provided here, are likely to facilitate further progress on the biology of versican and its proteolysis.
Collapse
|
8
|
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
|
9
|
Mutoji KN, Sun M, Elliott G, Moreno IY, Hughes C, Gesteira TF, Coulson-Thomas VJ. Extracellular Matrix Deposition and Remodeling after Corneal Alkali Burn in Mice. Int J Mol Sci 2021; 22:5708. [PMID: 34071909 PMCID: PMC8199272 DOI: 10.3390/ijms22115708] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022] Open
Abstract
Corneal transparency relies on the precise arrangement and orientation of collagen fibrils, made of mostly Type I and V collagen fibrils and proteoglycans (PGs). PGs are essential for correct collagen fibrillogenesis and maintaining corneal homeostasis. We investigated the spatial and temporal distribution of glycosaminoglycans (GAGs) and PGs after a chemical injury. The chemical composition of chondroitin sulfate (CS)/dermatan sulfate (DS) and heparan sulfate (HS) were characterized in mouse corneas 5 and 14 days after alkali burn (AB), and compared to uninjured corneas. The expression profile and corneal distribution of CS/DSPGs and keratan sulfate (KS) PGs were also analyzed. We found a significant overall increase in CS after AB, with an increase in sulfated forms of CS and a decrease in lesser sulfated forms of CS. Expression of the CSPGs biglycan and versican was increased after AB, while decorin expression was decreased. We also found an increase in KS expression 14 days after AB, with an increase in lumican and mimecan expression, and a decrease in keratocan expression. No significant changes in HS composition were noted after AB. Taken together, our study reveals significant changes in the composition of the extracellular matrix following a corneal chemical injury.
Collapse
Affiliation(s)
- Kazadi N. Mutoji
- College of Optometry, University of Houston, Houston, TX 77204, USA; (K.N.M.); (M.S.); (G.E.); (I.Y.M.); (T.F.G.)
| | - Mingxia Sun
- College of Optometry, University of Houston, Houston, TX 77204, USA; (K.N.M.); (M.S.); (G.E.); (I.Y.M.); (T.F.G.)
| | - Garrett Elliott
- College of Optometry, University of Houston, Houston, TX 77204, USA; (K.N.M.); (M.S.); (G.E.); (I.Y.M.); (T.F.G.)
| | - Isabel Y. Moreno
- College of Optometry, University of Houston, Houston, TX 77204, USA; (K.N.M.); (M.S.); (G.E.); (I.Y.M.); (T.F.G.)
| | - Clare Hughes
- School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK;
| | - Tarsis F. Gesteira
- College of Optometry, University of Houston, Houston, TX 77204, USA; (K.N.M.); (M.S.); (G.E.); (I.Y.M.); (T.F.G.)
- Optimvia, Batavia, OH 45103, USA
| | - Vivien J. Coulson-Thomas
- College of Optometry, University of Houston, Houston, TX 77204, USA; (K.N.M.); (M.S.); (G.E.); (I.Y.M.); (T.F.G.)
| |
Collapse
|
10
|
Tayman MA, Koyuncu İ, Köklü NÖ. Expression Levels of A Disintegrin-like Metalloproteinase with Thrombospondin Motifs-4 and -5 (ADAMTS-4 and ADAMTS-5) in Inflamed and Healthy Gingival Tissues. Comb Chem High Throughput Screen 2021; 23:168-176. [PMID: 32067610 DOI: 10.2174/1386207323666200218113000] [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: 08/30/2019] [Revised: 12/03/2019] [Accepted: 01/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND ADAMTS (A disintegrin-like metalloproteinase with thrombospondin motifs) is a group of 19 zinc-dependent metalloproteases known to function in many pathological and physiological processes, such as adhesion, cell fusion, signaling, proteolysis and ECM degradation. OBJECTIVES The aim of this study was to demonstrate the levels of ADAMTS-4 and -5 in gingival tissues with Stage III-Grade B generalized periodontitis (SIII-GB), Stage III-Grade C generalized periodontitis (SIII-GC) and healthy-control (C) groups. METHODS The clinical measurements were recorded for each patient. A total of 63 gingival biopsy specimens were obtained from the C (n:20), SIII-GB (n:23) and SIII-GC (n:20) groups. Polymerase chain reaction (Rt-PCR) and immunohistochemical (IHC) examinations were used to determine gene and protein levels. RESULTS According to the results of all methods, ADAMTS-4 and -5 expressions existed in periodontitis and C groups (P> 0.05). Immunostaining for ADAMTS-4 was found to be higher in patients with periodontitis than for ADAMTS-5 (P>0.05). Gene expression levels for ADAMTS-4 and -5 seemed to be up-regulated in subjects diagnosed with periodontitis, but the results were not statistically significant (P>0.05). A positive correlation was observed between PPD and ADAMTS-4 mRNA in SIII-GC (p=0.035) and SIII-GB (p=0.015). A positive correlation was determined between ADAMTS-4 mRNA and ADAMTS-5 mRNA in SIII-GC (p=0.037) and SIII-GB (p=0.00). CONCLUSION ADAMTS expression may take part in both pathological and physiological processes in the periodontal tissues, and periodontal destruction may be the result of a complex interaction of several pathways with many participants, such as ADAMTS-4 and -5, thus facilitating the exaggeration of periodontal disease.
Collapse
Affiliation(s)
- Mahmure A Tayman
- Department of Periodontology, Faculty of Dentistry, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - İsmail Koyuncu
- Department of Biochemistry, Faculty of Medicine, Harran University, Şanlıurfa, Turkey
| | - Nimet Ö Köklü
- Department of Pathology, Zekai Tahir Burak, Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
11
|
Kleiser S, Nyström A. Interplay between Cell-Surface Receptors and Extracellular Matrix in Skin. Biomolecules 2020; 10:E1170. [PMID: 32796709 PMCID: PMC7465455 DOI: 10.3390/biom10081170] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
Skin consists of the epidermis and dermis, which are connected by a specialized basement membrane-the epidermal basement membrane. Both the epidermal basement membrane and the underlying interstitial extracellular matrix (ECM) created by dermal fibroblasts contain distinct network-forming macromolecules. These matrices play various roles in order to maintain skin homeostasis and integrity. Within this complex interplay of cells and matrices, cell surface receptors play essential roles not only for inside-out and outside-in signaling, but also for establishing mechanical and biochemical properties of skin. Already minor modulations of this multifactorial cross-talk can lead to severe and systemic diseases. In this review, major epidermal and dermal cell surface receptors will be addressed with respect to their interactions with matrix components as well as their roles in fibrotic, inflammatory or tumorigenic skin diseases.
Collapse
Affiliation(s)
- Svenja Kleiser
- Department of Dermatology, Faculty of Medicine and Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine and Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany
| |
Collapse
|
12
|
Addition of High Molecular Weight Hyaluronic Acid to Fibroblast-Like Stromal Cells Modulates Endogenous Hyaluronic Acid Metabolism and Enhances Proteolytic Processing and Secretion of Versican. Cells 2020; 9:cells9071681. [PMID: 32668663 PMCID: PMC7407811 DOI: 10.3390/cells9071681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
We have examined the effect of exogenous linear chain high molecular weight hyaluronic acid (HMW HA) on endogenously synthesized hyaluronic acid (HA) and associated binding proteins in primary cultures of fibroblast-like stromal cells that were obtained by collagenase digestion of the murine peripatellar fat pad. The cultures were expanded in DMEM that was supplemented with fetal bovine serum and basic fibroblast growth factor (bFGF) then exposed to macrophage-colony-stimulating factor (MCSF) to induce macrophage properties, before activation of inflammatory pathways using E. coli lipopolysaccharide (LPS). Under all culture conditions, a significant amount of endogenously synthesized HA localized in LAMP1-positive lysosomal vesicles. However, this intracellular pool was depleted after the addition of exogenous HMW HA and was accompanied by enhanced proteolytic processing and secretion of de novo synthesized versican, much of which was associated with endosomal compartments. No changes were detected in synthesis, secretion, or proteolytic processing of aggrecan or lubricin (PRG4). The addition of HMW HA also modulated a range of LPS-affected genes in the TLR signaling and phagocytosis pathways, as well as endogenous HA metabolism genes, such as Has1, Hyal1, Hyal2, and Tmem2. However, there was no evidence for association of endogenous or exogenous HMW HA with cell surface CD44, TLR2 or TLR4 protein, suggesting that its physiochemical effects on pericelluar pH and/or ionic strength might be the primary modulators of signal transduction and vesicular trafficking by this cell type. We discuss the implications of these findings in terms of a potential in vivo effect of therapeutically applied HMW HA on the modification of osteoarthritis-related joint pathologies, such as pro-inflammatory and degradative responses of multipotent mesenchymal cells residing in the synovial membrane, the underlying adipose tissue, and the articular cartilage surface.
Collapse
|
13
|
Santamaria S. ADAMTS-5: A difficult teenager turning 20. Int J Exp Pathol 2020; 101:4-20. [PMID: 32219922 DOI: 10.1111/iep.12344] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/28/2019] [Accepted: 01/19/2020] [Indexed: 12/13/2022] Open
Abstract
A Disintegrin And Metalloproteinase with ThromboSpondin motif (ADAMTS)-5 was identified in 1999 as one of the enzymes responsible for cleaving aggrecan, the major proteoglycan in articular cartilage. Studies in vitro, ex vivo and in vivo have validated ADAMTS-5 as a target in osteoarthritis (OA), a disease characterized by extensive degradation of aggrecan. For this reason, it attracted the interest of many research groups aiming to develop a therapeutic treatment for OA patients. However, ADAMTS-5 proteoglycanase activity is not only involved in the dysregulated aggrecan proteolysis, which occurs in OA, but also in the physiological turnover of other related proteoglycans. In particular, versican, a major ADAMTS-5 substrate, plays an important structural role in heart and blood vessels and its proteolytic processing by ADAMTS-5 must be tightly regulated. On the occasion of the 20th anniversary of the discovery of ADAMTS-5, this review looks at the evidence for its detrimental role in OA, as well as its physiological turnover of cardiovascular proteoglycans. Moreover, the other potential functions of this enzyme are highlighted. Finally, challenges and emerging trends in ADAMTS-5 research are discussed.
Collapse
|
14
|
Pang X, Dong N, Zheng Z. Small Leucine-Rich Proteoglycans in Skin Wound Healing. Front Pharmacol 2020; 10:1649. [PMID: 32063855 PMCID: PMC6997777 DOI: 10.3389/fphar.2019.01649] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Healing of cutaneous wounds is a complex and well-coordinated process requiring cooperation among multiple cells from different lineages and delicately orchestrated signaling transduction of a diversity of growth factors, cytokines, and extracellular matrix (ECM) at the wound site. Most skin wound healing in adults is imperfect, characterized by scar formation which results in significant functional and psychological sequelae. Thus, the reconstruction of the damaged skin to its original state is of concern to doctors and scientists. Beyond the traditional treatments such as corticosteroid injection and radiation therapy, several growth factors or cytokines-based anti-scarring products are being or have been tested in clinical trials to optimize skin wound healing. Unfortunately, all have been unsatisfactory to date. Currently, accumulating evidence suggests that the ECM not only functions as the structural component of the tissue but also actively modulates signal transduction and regulates cellular behaviors, and thus, ECM should be considered as an alternative target for wound management pharmacotherapy. Of particular interest are small leucine-rich proteoglycans (SLRPs), a group of the ECM, which exist in a wide range of connecting tissues, including the skin. This manuscript summarizes the most current knowledge of SLRPs regarding their spatial-temporal expression in the skin, as well as lessons learned from the genetically modified animal models simulating human skin pathologies. In this review, particular focus is given on the diverse roles of SLRP in skin wound healing, such as anti-inflammation, pro-angiogenesis, pro-migration, pro-contraction, and orchestrate transforming growth factor (TGF)β signal transduction, since cumulative investigations have indicated their therapeutic potential on reducing scar formation in cutaneous wounds. By conducting this review, we intend to gain insight into the potential application of SLRPs in cutaneous wound healing management which may pave the way for the development of a new generation of pharmaceuticals to benefit the patients suffering from skin wounds and their sequelae.
Collapse
Affiliation(s)
- Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nuo Dong
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Rezvani SN, Chen J, Li J, Midura R, Cali V, Sandy JD, Plaas A, Wang VM. In-Vivo Efficacy of Recombinant Human Hyaluronidase (rHuPH20) Injection for Accelerated Healing of Murine Retrocalcaneal Bursitis and Tendinopathy. J Orthop Res 2020; 38:59-69. [PMID: 31478241 PMCID: PMC6917826 DOI: 10.1002/jor.24459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
The deposition of aggrecan/hyaluronan (HA)-rich matrix within the tendon body and surrounding peritenon impede tendon healing and result in compromised biomechanical properties. Hence, the development of novel strategies to achieve targeted removal of the aggrecan-HA pericellular matrix may be effective in treating tendinopathy. The current study examined the therapeutic potential of a recombinant human hyaluronidase, rHuPH20 (FDA approved for reducing HA accumulation in tumors) for treating murine Achilles tendinopathy. The 12-week-old C57Bl/6 male mice were injected with two doses of rHuTGF-β1 into the retrocalcaneal bursa (RCB) to induce a combined bursitis and tendinopathy. Twenty-four hours following induction of injury, treatment groups were administered rHuPH20 Hyaluronidase (rHuPH20; Halozyme Therapeutics) into the RCB. At either 6 h (acute), 9 days, or 25 days following hyaluronidase treatment, Achilles tendons were analyzed for gene expression, histology and immunohistochemistry, fluorophore-assisted carbohydrate electrophoresis, and biomechanical properties. The rHuPH20 treatment was effective, particularly at the acute and 9-day time points, in (a) removing HA deposits from the Achilles tendon and surrounding tissues, (b) improving biomechanical properties of the healing tendon, and (c) eliciting targeted increases in expression of specific cell fate, extracellular matrix metabolism, and inflammatory genes. The potential of rHuPH20 to effectively clear the pro-inflammatory, HA-rich matrix within the RCB and tendon strongly supports the future refinement of injectable glycosidase preparations as potential treatments to protect or regenerate tendon tissue by reducing inflammation and scarring in the presence of bursitis or other inducers of damage such as mechanical overuse. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:59-69, 2020.
Collapse
Affiliation(s)
- Sabah N. Rezvani
- Department of Biomedical Engineering and Mechanics, Virginia Tech (Blacksburg, VA)
| | - Jinnan Chen
- Department of Internal Medicine (Rheumatology), Rush University (Chicago, IL)
| | - Jun Li
- Department of Internal Medicine (Rheumatology), Rush University (Chicago, IL)
| | - Ron Midura
- Lerner Research Institute, The Cleveland Clinic Foundation (Cleveland, Ohio)
| | - Valbona Cali
- Lerner Research Institute, The Cleveland Clinic Foundation (Cleveland, Ohio)
| | - John D. Sandy
- Department of Orthopedic Surgery, Rush University (Chicago, IL)
| | - Anna Plaas
- Department of Internal Medicine (Rheumatology), Rush University (Chicago, IL)
| | - Vincent M. Wang
- Department of Biomedical Engineering and Mechanics, Virginia Tech (Blacksburg, VA)
| |
Collapse
|
16
|
Islam S, Chuensirikulchai K, Khummuang S, Keratibumrungpong T, Kongtawelert P, Kasinrerk W, Hatano S, Nagamachi A, Honda H, Watanabe H. Accumulation of versican facilitates wound healing: Implication of its initial ADAMTS-cleavage site. Matrix Biol 2019; 87:77-93. [PMID: 31669737 DOI: 10.1016/j.matbio.2019.10.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022]
Abstract
Versican is a large chondroitin sulfate/dermatan sulfate proteoglycan in the extracellular matrix, and is expressed at high levels in tissues during development and remodeling in pathological conditions. Its core protein is cleaved at a region close to the N-terminal end of CSβ domain by several members of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, i.e., ADAMTS-1, 4, 5, 9, 15, and 20. Here, using a CRISPR/Cas9 system, we generated knock-in mice (V1R), which express an ADAMTS cleavage-resistant versican. Some V1R homozygote mice, termed R/R, exhibit syndactyly and organ hemorrhage. In wound healing experiments, R/R wound shows accumulation of versican and activated TGFβ-signaling in the early stage, leading to faster healing than wild type wound. Immunostaining for Ki67, CD31, smooth muscle α-actin, periostin demonstrates higher levels of overall cell proliferation and an increased number of endothelial cells and myofibroblasts. Immunostaining for CD11b and qRT-PCR for macrophage markers revealed increased levels of inflammatory cell infiltration, especially those of M1 macrophages. Cultured R/R dermal fibroblasts revealed increased deposition of versican, type I and III collagens, and hyaluronan, and upregulation of Smad2/3 signaling. Taken together, these results demonstrate that the cleavage site determines versican turnover and that versican plays a central role in the provisional matrix during the wound repair.
Collapse
Affiliation(s)
- Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Kantinan Chuensirikulchai
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Biomedical Technology Research Center, Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Saichit Khummuang
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Biomedical Technology Research Center, Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Tanyaporn Keratibumrungpong
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Watchara Kasinrerk
- Biomedical Technology Research Center, Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sonoko Hatano
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Akiko Nagamachi
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan.
| |
Collapse
|
17
|
Wang Y, Mack JA, Maytin EV. CD44 inhibits α-SMA gene expression via a novel G-actin/MRTF-mediated pathway that intersects with TGFβR/p38MAPK signaling in murine skin fibroblasts. J Biol Chem 2019; 294:12779-12794. [PMID: 31285260 DOI: 10.1074/jbc.ra119.007834] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/25/2019] [Indexed: 01/10/2023] Open
Abstract
Well-regulated differentiation of fibroblasts into myofibroblasts (MF) is critical for skin wound healing. Neoexpression of α-smooth muscle actin (α-SMA), an established marker for MF differentiation, is driven by TGFβ receptor (TGFβR)-mediated signaling. Hyaluronan (HA) and its receptor CD44 may also participate in this process. To further understand this process, primary mouse skin fibroblasts were isolated and treated in vitro with recombinant TGF-β1 (rTGF-β1) to induce α-SMA expression. CD44 expression was also increased. Paradoxically, CD44 knockdown by RNA interference (RNAi) led to increased α-SMA expression and α-SMA-containing stress fibers. Removal of extracellular HA or inhibition of HA synthesis had no effect on α-SMA levels, suggesting a dispensable role for HA. Exploration of mechanisms linking CD44 knockdown to α-SMA induction, using RNAi and chemical inhibitors, revealed a requirement for noncanonical TGFβR signaling through p38MAPK. Decreased monomeric G-actin but increased filamentous F-actin following CD44 RNAi suggested a possible role for myocardin-related transcription factor (MRTF), a known regulator of α-SMA transcription and itself regulated by G-actin binding. CD44 RNAi promoted nuclear accumulation of MRTF and the binding to its transcriptional cofactor SRF. MRTF knockdown abrogated the increased α-SMA expression caused by CD44 RNAi, suggesting that MRTF is required for CD44-mediated regulation of α-SMA. Finally, chemical inhibition of p38MAPK reversed nuclear MRTF accumulation after rTGF-β1 addition or CD44 RNAi, revealing a central involvement of p38MAPK in both cases. We concluded that CD44 regulates α-SMA gene expression through cooperation between two intersecting signaling pathways, one mediated by G-actin/MRTF and the other via TGFβR/p38MAPK.
Collapse
Affiliation(s)
- Yan Wang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Judith A Mack
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Edward V Maytin
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195 .,Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
18
|
The role of kinin B 1 and B 2 receptors in the mouse model of oxazolone-induced atopic dermatitis. Int Immunopharmacol 2019; 72:62-73. [PMID: 30959373 DOI: 10.1016/j.intimp.2019.03.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/23/2019] [Accepted: 03/29/2019] [Indexed: 01/01/2023]
Abstract
This study evaluated the role of kinin B1 and B2 receptors in the pre-clinical mouse model of oxazolone-induced atopic dermatitis. The B1 R715 or B2 HOE140 receptor antagonists were dosed at different schemes of treatment. After assessment of clinical lesion scores and pruritus, lesional skin samples were collected for histopathological analysis. The plasma extravasation and the expression of the metalloproteinase ADAMTS5 were also assessed. The immunopositivity for kinin receptors was evaluated in the skin, dorsal root ganglion (DRG), thoracic spinal cord and brain cortex sections. Marked upregulation of B1 and B2 receptors was observed in the skin of oxazolone-treated mice. The induction of atopic dermatitis led to a downregulation of both receptors in the DRG, without any alteration in the spinal cord and brain cortex. The repeated administration of HOE140 (50 nmol/kg; i.p.) partially inhibited the oxazolone-related pruritus, associated with a reduction of ADAMTS5 immunolabelling in the skin. Alternatively, R715 (438 nmol/kg; i.p.) produced a mild inhibition of plasma extravasation in oxazolone-challenged mice. Noteworthy, the repeated i.d. injection of R715 (30 nmol/site) or HOE140 (3 nmol/site) significantly reduced the histiocyte numbers, according to the histopathological analysis. Either B1 or B2 kinin antagonists, irrespective of the protocol of treatment, did not alter any other evaluated clinical or histological parameters. Data brings novel evidence about the role of kinin receptors in allergy-related conditions, such as atopic dermatitis. Further studies to test different protocols of treatment with kinin antagonists on in-depth cellular alterations underlying oxazolone-induced atopic dermatitis remain to be performed.
Collapse
|
19
|
Sikes KJ, Li J, Shen Q, Gao SG, Sandy JD, Plaas A, Wang VM. TGF-b1 or hypoxia enhance glucose metabolism and lactate production via HIF1A signaling in tendon cells. Connect Tissue Res 2018; 59:458-471. [PMID: 29447016 PMCID: PMC6175639 DOI: 10.1080/03008207.2018.1439483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
UNLABELLED Purpose/Aim of the study: Healthy tendons are maintained in homeostasis through controlled usage of glucose for energy and redox equilibrium. Tendon cell stress imposed by overuse injury or vascular insufficiency is accompanied by activation of wound healing pathways which facilitate an adaptive response and the restoration of homeostasis. To understand this response at the gene expression level we have studied the in vivo effects of injected TGF-β1 in a murine model of tendinopathy, as well as treatment of murine tendon explants with either TGF-β1 or hypoxia in vitro. METHODS AND RESULTS We provide evidence (from expression patterns and immunohistochemistry) that both in vivo and in vitro, the stress response in tendon cells may be metabolically controlled in part by glycolytic reprogramming. A major feature of the response to TGF-β1 or hypoxia is activation of the Warburg pathway which generates lactate from glucose under normoxia and thereby inhibits mitochondrial energy production. CONCLUSIONS We discuss the likely outcome of this major metabolic shift in terms of the potential benefits and damage to tendon and suggest how incorporation of this metabolic response into our understanding of initiation and progression of tendinopathies may offer new opportunities for diagnosis and the monitoring of therapies.
Collapse
Affiliation(s)
- Katie J Sikes
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, Chicago, IL 60607
| | - Jun Li
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Quan Shen
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China 410008
| | - Shu-Guang Gao
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China 410008
| | - John D Sandy
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Anna Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Vincent M Wang
- Department of Biomedical Engineering and Mechanics, Virginia Tech, 339 Kelly Hall, 325 Stanger Street MC 0298, Blacksburg, VA, 24061,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, Chicago, IL 60607
| |
Collapse
|
20
|
Expression of ADAMTS2 and ADAMTS5 in the salivary gland of rats after radioiodine therapy. Nucl Med Commun 2018; 39:110-117. [PMID: 29194287 DOI: 10.1097/mnm.0000000000000781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the presence of ADAMTS2 and ADAMTS5 in the salivary gland (SG) of rats after high-dose radioiodine therapy. METHODS A total of 36 male Wistar albino rats were used for this study. Thirty-six male rats were divided randomly into six groups: control and five radioactive iodine (RAI) treatment groups of six rats each. All animals were killed. The evaluation of biodistribution and histopathological studies were carried out on the SGs removed. Real-time PCR and immunohistochemical analysis were carried out to determine mRNA and protein expression levels of ADAMTS genes. Differences between the groups were evaluated statistically. RESULTS In RAI-treated groups, ADAMTS2 and ADAMTS5 gene expression was observed to increase, whereas there was no mRNA or protein expression in the control group. There were statistically significant increases in the mRNA expression of ADAMTS2 (all RAI-administered groups in parathyroid gland and at 4, 24, and 48 h in submandibular gland) and ADAMTS5 (all RAI-administered groups, except on the 30th day in the parathyroid gland and all RAI groups in submandibular gland). Through immunohistochemical analysis, the staining pattern in the extracellular source was also observed in the overexpressed ADAMTS2 and ADAMTS5 groups. Nuclear coarsening and partial focal subnuclei vacuolization were determined in all RAI-administered groups with histopathological examinations. CONCLUSION An increase in the mRNA expression levels of ADAMTS2 and ADAMTS5 genes was detected in the RAI-administered groups. These results suggested that ADAMTS2 and ADAMTS5 genes might play a role in radiation exposure and radioiodine-induced SG changes.
Collapse
|
21
|
Ricard-Blum S, Baffet G, Théret N. Molecular and tissue alterations of collagens in fibrosis. Matrix Biol 2018; 68-69:122-149. [DOI: 10.1016/j.matbio.2018.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
|
22
|
Perrucci GL, Rurali E, Pompilio G. Cardiac fibrosis in regenerative medicine: destroy to rebuild. J Thorac Dis 2018; 10:S2376-S2389. [PMID: 30123577 DOI: 10.21037/jtd.2018.03.82] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The major limitations for cardiac regeneration in patients after myocardial infarction (MI) are the wide loss of cardiomyocytes and the adverse structural alterations of extracellular matrix (ECM). Cardiac fibroblast differentiation into myofibroblasts (MFB) leads to a huge deposition of ECM and to the subsequent loss of ventricular structural integrity. All these molecular events depict the fundamental features at the basis of the post-MI fibrosis and deserve in depth cellular and molecular studies to fill the gap in the clinical practice. Indeed, to date, there are no effective therapeutic approaches to limit the post-MI massive fibrosis development. In this review we describe the involvement of integrins and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)/ADAMTS-like (ADAMTSL) proteins in cardiac reparative pro-fibrotic response after MI, proposing some of them as novel potential pharmacological tools.
Collapse
Affiliation(s)
- Gianluca Lorenzo Perrucci
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Milano, Italy.,Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino-IRCCS, Milano, Italy
| | - Erica Rurali
- Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino-IRCCS, Milano, Italy
| | - Giulio Pompilio
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Milano, Italy.,Unità di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino-IRCCS, Milano, Italy.,Dipartimento di Chirurgia Cardiovascolare, Centro Cardiologico Monzino-IRCCS, Milano, Italy
| |
Collapse
|
23
|
Piperigkou Z, Götte M, Theocharis AD, Karamanos NK. Insights into the key roles of epigenetics in matrix macromolecules-associated wound healing. Adv Drug Deliv Rev 2018; 129:16-36. [PMID: 29079535 DOI: 10.1016/j.addr.2017.10.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/14/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
Abstract
Extracellular matrix (ECM) is a dynamic network of macromolecules, playing a regulatory role in cell functions, tissue regeneration and remodeling. Wound healing is a tissue repair process necessary for the maintenance of the functionality of tissues and organs. This highly orchestrated process is divided into four temporally overlapping phases, including hemostasis, inflammation, proliferation and tissue remodeling. The dynamic interplay between ECM and resident cells exerts its critical role in many aspects of wound healing, including cell proliferation, migration, differentiation, survival, matrix degradation and biosynthesis. Several epigenetic regulatory factors, such as the endogenous non-coding microRNAs (miRNAs), are the drivers of the wound healing response. microRNAs have pivotal roles in regulating ECM composition during wound healing and dermal regeneration. Their expression is associated with the distinct phases of wound healing and they serve as target biomarkers and targets for systematic regulation of wound repair. In this article we critically present the importance of epigenetics with particular emphasis on miRNAs regulating ECM components (i.e. glycoproteins, proteoglycans and matrix proteases) that are key players in wound healing. The clinical relevance of miRNA targeting as well as the delivery strategies designed for clinical applications are also presented and discussed.
Collapse
|
24
|
Mahale A, Fikri F, Al Hati K, Al Shahwan S, Al Jadaan I, Al Katan H, Khandekar R, Maktabi A, Edward DP. Histopathologic and immunohistochemical features of capsular tissue around failed Ahmed glaucoma valves. PLoS One 2017; 12:e0187506. [PMID: 29121102 PMCID: PMC5679546 DOI: 10.1371/journal.pone.0187506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/20/2017] [Indexed: 01/03/2023] Open
Abstract
Impervious encapsulation around Ahmed glaucoma valve (AGV) results in surgical failure raising intraocular pressure (IOP). Dysregulation of extracellular matrix (ECM) molecules and cellular factors might contribute to increased hydraulic resistance to aqueous drainage. Therefore, we examined these molecules in failed AGV capsular tissue. Immunostaining for ECM molecules (collagen I, collagen III, decorin, lumican, chondroitin sulfate, aggrecan and keratan sulfate) and cellular factors (αSMA and TGFβ) was performed on excised capsules from failed AGVs and control tenon’s tissue. Staining intensity of ECM molecules was assessed using Image J. Cellular factors were assessed based on positive cell counts. Histopathologically two distinct layers were visible in capsules. The inner layer (proximal to the AGV) showed significant decrease in most ECM molecules compared to outer layer. Furthermore, collagen III (p = 0.004), decorin (p = 0.02), lumican (p = 0.01) and chondroitin sulfate (p = 0.02) was significantly less in inner layer compared to tenon’s tissue. Outer layer labelling however was similar to control tenon’s for most ECM molecules. Significantly increased cellular expression of αSMA (p = 0.02) and TGFβ (p = 0.008) was detected within capsular tissue compared to controls. Our results suggest profibrotic activity indicated by increased αSMA and TGFβ expression and decreased expression of proteoglycan (decorin and lumican) and glycosaminoglycans (chondroitin sulfate). Additionally, we observed decreased collagen III which might reflect increased myofibroblast contractility when coupled with increased TGFβ and αSMA expression. Together these events lead to tissue dysfunction potentially resulting in hydraulic resistance that may affect aqueous flow through the capsular wall.
Collapse
Affiliation(s)
- Alka Mahale
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Fatma Fikri
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Khitam Al Hati
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Sami Al Shahwan
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Ibrahim Al Jadaan
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Hind Al Katan
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Rajiv Khandekar
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Azza Maktabi
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Deepak P. Edward
- King Khaled Eye Specialist Hospital, Riyadh, Kingdom of Saudi Arabia
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail: ,
| |
Collapse
|
25
|
The ADAMTS hyalectanase family: biological insights from diverse species. Biochem J 2017; 473:2011-22. [PMID: 27407170 DOI: 10.1042/bcj20160148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/29/2016] [Indexed: 12/13/2022]
Abstract
The a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs (ADAMTS) family of metzincins are complex secreted proteins that have diverse functions during development. The hyalectanases (ADAMTS1, 4, 5, 8, 9, 15 and 20) are a subset of this family that have enzymatic activity against hyalectan proteoglycans, the processing of which has important implications during development. This review explores the evolution, expression and developmental functions of the ADAMTS family, focusing on the ADAMTS hyalectanases and their substrates in diverse species. This review gives an overview of how the family and their substrates evolved from non-vertebrates to mammals, the expression of the hyalectanases and substrates in different species and their functions during development, and how these functions are conserved across species.
Collapse
|
26
|
Trella KJ, Li J, Stylianou E, Wang VM, Frank JM, Galante J, Sandy JD, Plaas A, Wysocki R. Genome-wide analysis identifies differential promoter methylation of Leprel2, Foxf1, Mmp25, Igfbp6, and Peg12 in murine tendinopathy. J Orthop Res 2017; 35:947-955. [PMID: 27517731 PMCID: PMC5303683 DOI: 10.1002/jor.23393] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/09/2016] [Indexed: 02/04/2023]
Abstract
We have used a murine Achilles tendinopathy model to investigate whether tissue changes (such as collagen disorganization, chondroid metaplasia, and loss of tensile properties) which are broadly characteristic of human tendinopathies, are accompanied by changes in the expression of chromatin-modifying enzymes and the methylation status of promoter regions of tendon cell DNA. Tendinopathy was induced by two intra-tendinous TGF-β1 injections followed by cage activity or treadmill running for up to 28 days. Activation of DNA methyltransferases occurred at 3 days after the TGF-β1 injections and also at 14 days, but only with treadmill activity. Genome-wide Methyl Mini-Seq™ analysis identified 19 genes with differentially methylated promoters, five of which perform functions with an apparent direct relevance to tendinopathy (Leprel2, Foxf1, Mmp25, Igfbp6, and Peg12). The functions of the genes identified included collagen fiber assembly and pericellular interactions, therefore their perturbation could play a role in the characteristic disorganization of fibers in affected tendons. We postulate that a study of the functional genomics of these genes in animal and human tendon could further delineate the pathogenesis of this multi-factorial complex disease. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:947-955, 2017.
Collapse
Affiliation(s)
- Katie J Trella
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 201, Chicago, IL 60612,Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, 2 Floor, Chicago, IL 60607
| | - Jun Li
- Department of Rheumatology/Internal Medicine, Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612
| | - Eleni Stylianou
- Formerly Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195
| | - Vincent M Wang
- Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, 2 Floor, Chicago, IL 60607,Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061
| | - Jonathan M Frank
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 201, Chicago, IL 60612
| | - Jorge Galante
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 201, Chicago, IL 60612
| | - John D Sandy
- Department of Biochemistry, Rush University Medical Center, 1735 W. Harrison Street, 5 Floor, Chicago, IL 60612
| | - Anna Plaas
- Department of Rheumatology/Internal Medicine, Rush University Medical Center, 1611 W. Harrison Street, Suite 510, Chicago, IL 60612,Department of Biochemistry, Rush University Medical Center, 1735 W. Harrison Street, 5 Floor, Chicago, IL 60612
| | - Robert Wysocki
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 201, Chicago, IL 60612
| |
Collapse
|
27
|
Thurairajah K, Broadhead ML, Balogh ZJ. Trauma and Stem Cells: Biology and Potential Therapeutic Implications. Int J Mol Sci 2017; 18:ijms18030577. [PMID: 28272352 PMCID: PMC5372593 DOI: 10.3390/ijms18030577] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/11/2022] Open
Abstract
Trauma may cause irreversible tissue damage and loss of function despite current best practice. Healing is dependent both on the nature of the injury and the intrinsic biological capacity of those tissues for healing. Preclinical research has highlighted stem cell therapy as a potential avenue for improving outcomes for injuries with poor healing capacity. Additionally, trauma activates the immune system and alters stem cell behaviour. This paper reviews the current literature on stem cells and its relevance to trauma care. Emphasis is placed on understanding how stem cells respond to trauma and pertinent mechanisms that can be utilised to promote tissue healing. Research involving notable difficulties in trauma care such as fracture non-union, cartilage damage and trauma induced inflammation is discussed further.
Collapse
Affiliation(s)
- Kabilan Thurairajah
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Department of Traumatology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia.
| | - Matthew L Broadhead
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Department of Traumatology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia.
| | - Zsolt J Balogh
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Department of Traumatology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
28
|
Degenerative Suspensory Ligament Desmitis (DSLD) in Peruvian Paso Horses Is Characterized by Altered Expression of TGFβ Signaling Components in Adipose-Derived Stromal Fibroblasts. PLoS One 2016; 11:e0167069. [PMID: 27902739 PMCID: PMC5130251 DOI: 10.1371/journal.pone.0167069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/08/2016] [Indexed: 01/06/2023] Open
Abstract
Equine degenerative suspensory ligament desmitis (DSLD) in Peruvian Paso horses typically presents at 7–15 years and is characterized by lameness, focal disorganization of collagen fibrils, and chondroid deposition in the body of the ligament. With the aim of developing a test for disease risk (that can be used to screen horses before breeding) we have quantified the expression of 76 TGFβ-signaling target genes in adipose-derived stromal fibroblasts (ADSCs) from six DSLD-affected and five unaffected Paso horses. Remarkably, 35 of the genes showed lower expression (p<0.05) in cells from DSLD-affected animals and this differential was largely eliminated by addition of exogenous TGFβ1. Moreover, TGFβ1-mediated effects on expression were prevented by the TGFβR1/2 inhibitor LY2109761, showing that the signaling was via a TGFβR1/2 complex. The genes affected by the pathology indicate that it is associated with a generalized metabolic disturbance, since some of those most markedly altered in DSLD cells (ATF3, MAPK14, ACVRL1 (ALK1), SMAD6, FOS, CREBBP, NFKBIA, and TGFBR2) represent master-regulators in a wide range of cellular metabolic responses.
Collapse
|
29
|
Gurses MS, Ural MN, Gulec MA, Akyol O, Akyol S. Pathophysiological Function of ADAMTS Enzymes on Molecular Mechanism of Alzheimer's Disease. Aging Dis 2016; 7:479-90. [PMID: 27493839 PMCID: PMC4963191 DOI: 10.14336/ad.2016.0111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/11/2016] [Indexed: 12/17/2022] Open
Abstract
The extracellular matrix (ECM) is an environment that has various enzymes attended in regeneration and restoration processes which is very important to sustain physiological and biological functions of central nervous system (CNS). One of the participating enzyme systems in ECM turnover is matrix metalloproteinases. A disintegrin-like and metalloproteinase with thrombospondin type 1 motifs (ADAMTS) is a unique family of ECM proteases found in mammals. Components of this family may be distinguished from the ADAM (A Disintegrin and Metalloproteinase) family based on the multiple copies of thrombospondin 1-like repeats. The considerable role of the ADAMTS in the CNS continues to develop. Evidences indicate that ADAMTS play an important role in neuroplasticity as well as nervous system pathologies such as Alzheimer's disease (AD). It is hopeful and possible that ADAMTS family members may be utilized to develop therapies for CNS pathologies, ischemic injuries, neurodegenerative and neurological diseases. To understand and provide definitive data on ADAMTS to improve structural and functional recovery in CNS injury and diseases, this review aimed to enlighten the subject extensively to reach certain information on metalloproteinases and related molecules/enzymes. It will be interesting to examine how ADAMTS expression and action would affect the initiation/progression of above-mentioned clinical situations, especially AD.
Collapse
Affiliation(s)
- Murat Serdar Gurses
- Department of Forensic Medicine, School of Medicine, Uludag University, Bursa, Turkey
| | - Mustafa Numan Ural
- Department of Forensic Medicine, School of Medicine, Uludag University, Bursa, Turkey
| | - Mehmet Akif Gulec
- Department of Medical Biochemistry, Faculty of Medicine, Turgut Ozal University, Ankara, Turkey
| | - Omer Akyol
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Sumeyya Akyol
- Department of Medical Biology, Faculty of Medicine, Turgut Ozal University, Ankara, Turkey
| |
Collapse
|
30
|
Chang YH, Liu HW, Wu KC, Ding DC. Mesenchymal Stem Cells and Their Clinical Applications in Osteoarthritis. Cell Transplant 2016; 25:937-50. [DOI: 10.3727/096368915x690288] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis is a chronic degenerative joint disorder characterized by articular cartilage destruction and osteophyte formation. Chondrocytes in the matrix have a relatively slow turnover rate, and the tissue itself lacks a blood supply to support repair and remodeling. Researchers have evaluated the effectiveness of stem cell therapy and tissue engineering for treating osteoarthritis. All sources of stem cells, including embryonic, induced pluripotent, fetal, and adult stem cells, have potential use in stem cell therapy, which provides a permanent biological solution. Mesenchymal stem cells (MSCs) isolated from bone marrow, adipose tissue, and umbilical cord show considerable promise for use in cartilage repair. MSCs can be sourced from any or all joint tissues and can modulate the immune response. Additionally, MSCs can directly differentiate into chondrocytes under appropriate signal transduction. They also have immunosuppressive and anti-inflammatory paracrine effects. This article reviews the current clinical applications of MSCs and future directions of research in osteoarthritis.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Hwan-Wun Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Occupational Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Kun-Chi Wu
- Department of Orthopedics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Dah-Ching Ding
- Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
31
|
Anti-ADAMTS5 monoclonal antibodies: implications for aggrecanase inhibition in osteoarthritis. Biochem J 2016; 473:e1-4. [PMID: 26657033 DOI: 10.1042/bj20151072] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The extracellular matrix of articular cartilage is structurally specialized for efficient absorption of mechanical impact. In particular, giant aggregates of the large chondroitin sulfate proteoglycan, aggrecan, with the glycosaminoglycan, hyaluronan, allow cartilage to resist compressive load. Proteolysis of aggrecan by members of the proteinase family ADAMTS (A disintegrin-like and metalloproteinase domain with thrombospondin type 1 motif), was identified as an early step in the inexorable destruction of cartilage in osteoarthritis (OA). Of the investigated proteinases, ADAMTS5 has emerged as a principal mediator of aggrecan loss in OA, convincingly so in mouse models, and with high probability in humans. ADAMTS5 has a bipartite organization, comprising a proteinase domain and an ancillary domain containing exosites for interaction with aggrecan and other substrates. In a recent issue of this journal, Santamaria et al. characterized anti-ADAMTS5 monoclonal antibodies isolated from a phage display library. By blocking the catalytic site of the ADAMTS5 immunogen with a synthetic inhibitor, the authors of the paper biased selection of antibodies to the ancillary domain. This work, together with other antibodies targeting ADAMTS5, offers diverse, high-affinity and, as far as can be determined, selective aggrecanase inhibitors. Mapping of their epitopes provided novel insights into ADAMTS5 interactions with aggrecan. These monoclonal antibodies deserve continued investigation for potential arthritis therapy, although their successful use will require a comprehensive understanding of the physiological roles of ADAMTS5, and its regulation, intrinsic properties and intermolecular interactions.
Collapse
|
32
|
Aydos SE, Yukselten Y, Sunguroglu A, Demircan K, Aydos K. Role of ADAMTS1 and ADAMTS5 in male infertility. Andrologia 2016; 48:1113-1119. [PMID: 26888488 DOI: 10.1111/and.12547] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2015] [Indexed: 11/28/2022] Open
Abstract
The aim of this study was to investigate the relationship of infertility with metalloproteinases ADAMTS1 and ADAMTS5, which are known to be responsible for the degradation of extracellular matrix (ECM) proteins associated with many diseases. ECM is the noncellular component that provides structural and biochemical support to the surrounding cells required for tissue morphogenesis, differentiation and homoeostasis. Sixty infertile individuals and 10 healthy semen donors were included in this study. The infertile individuals were classified as normozoospermia (NS; n = 20), oligozoospermia (OS; n = 20), azoospermia (AS; n = 20) groups. ADAMTS1 and ADAMTS5 protein levels in semen were analysed by Western blot. ADAMTS1 protein level was 3.0-, 3.3- and 1.6-fold lower in the OS, AS and NS groups, respectively, than in the control group (P < 0.001). ADAMTS5 protein level was 3.2-, 2.7- and 1.4-fold lower in the OS, AS and NS groups, respectively, than in the control group (P < 0.001). Sperm count and sperm motility showed a negative correlation with the levels of ADAMTS1 and ADAMTS5 protein expression: r = -0.477, r = -0.470; and r = -0.332, r = -0.275 respectively (P < 0.001). In conclusion, ADAMTS1 and ADAMTS5 protein expressions in semen are significantly related with sperm production. It is very important to understand molecular function and organisation of ADAMTSs which will be significant in enlightening the process of spermatogenesis in male infertility.
Collapse
Affiliation(s)
- S E Aydos
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | - Y Yukselten
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | - A Sunguroglu
- Department of Medical Biology, School of Medicine, Ankara University, Ankara, Turkey
| | - K Demircan
- Department of Medical Biology, School of Medicine, Turgut Ozal University, Ankara, Turkey
| | - K Aydos
- Department of Urology, School of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
33
|
Sayegh ET, Sandy JD, Virk MS, Romeo AA, Wysocki RW, Galante JO, Trella KJ, Plaas A, Wang VM. Recent Scientific Advances Towards the Development of Tendon Healing Strategies. ACTA ACUST UNITED AC 2015; 4:128-143. [PMID: 26753125 DOI: 10.2174/2211542004666150713190231] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There exists a range of surgical and non-surgical approaches to the treatment of both acute and chronic tendon injuries. Despite surgical advances in the management of acute tears and increasing treatment options for tendinopathies, strategies frequently are unsuccessful, due to impaired mechanical properties of the treated tendon and/or a deficiency in progenitor cell activities. Hence, there is an urgent need for effective therapeutic strategies to augment intrinsic and/or surgical repair. Such approaches can benefit both tendinopathies and tendon tears which, due to their severity, appear to be irreversible or irreparable. Biologic therapies include the utilization of scaffolds as well as gene, growth factor, and cell delivery. These treatment modalities aim to provide mechanical durability or augment the biologic healing potential of the repaired tissue. Here, we review the emerging concepts and scientific evidence which provide a rationale for tissue engineering and regeneration strategies as well as discuss the clinical translation of recent innovations.
Collapse
Affiliation(s)
- Eli T Sayegh
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - John D Sandy
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612
| | - Mandeep S Virk
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Anthony A Romeo
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Robert W Wysocki
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Jorge O Galante
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Katie J Trella
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| | - Anna Plaas
- Department of Rheumatology/Internal Medicine, Rush University Medical Center, Chicago, IL 60612
| | - Vincent M Wang
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
34
|
Gorski DJ, Xiao W, Li J, Luo W, Lauer M, Kisiday J, Plaas A, Sandy J. Deletion of ADAMTS5 does not affect aggrecan or versican degradation but promotes glucose uptake and proteoglycan synthesis in murine adipose derived stromal cells. Matrix Biol 2015; 47:66-84. [DOI: 10.1016/j.matbio.2015.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 03/25/2015] [Accepted: 03/26/2015] [Indexed: 01/22/2023]
|
35
|
Antibody-based exosite inhibitors of ADAMTS-5 (aggrecanase-2). Biochem J 2015; 471:391-401. [PMID: 26303525 PMCID: PMC4613496 DOI: 10.1042/bj20150758] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/24/2015] [Indexed: 11/17/2022]
Abstract
We isolated four antibody-based exosite inhibitors of adamalysin-like metalloproteinases with thrombospondin (TS) motifs (ADAMTS)-5, a multi-domain metalloproteinase, from a phage display library. One of them binds to the spacer domain (Sp) and inhibits the enzyme action selectively on natural substrate proteoglycans, but not on peptides. Adamalysin-like metalloproteinases with thrombospondin (TS) motifs (ADAMTS)-5 is the multi-domain metalloproteinase that most potently degrades aggrecan proteoglycan in the cartilage and its activity is implicated in the development of osteoarthritis (OA). To generate specific exosite inhibitors for it, we screened a phage display antibody library in the presence of the zinc-chelating active site-directed inhibitor GM6001 (Ilomastat) and isolated four highly selective inhibitory antibodies. Two antibodies were mapped to react with exosites in the catalytic/disintegrin domains (Cat/Dis) of the enzyme, one in the TS domain and one in the spacer domain (Sp). The antibody reacting with the Sp blocked the enzyme action only when aggrecan or the Escherichia coli-expressed aggrecan core protein were substrates, but not against a peptide substrate. The study with this antibody revealed the importance of the Sp for effective aggrecanolytic activity of ADAMTS-5 and that this domain does not interact with sulfated glycosaminoglycans (GAGs) but with the protein moiety of the proteoglycan. An antibody directed against the Cat/Dis of ADAMTS-5 was effective in a cell-based model of aggrecan degradation; however, the anti-Sp antibody was ineffective. Western blot analysis of endogenous ADAMTS-5 expressed by human chondrocytes showed the presence largely of truncated forms of ADAMTS-5, thus explaining the lack of efficacy of the anti-Sp antibody. The possibility of ADAMTS-5 truncation must then be taken into account when considering developing anti-ancillary domain antibodies for therapeutic purposes.
Collapse
|
36
|
Similarities in Gene Expression Profiles during In Vitro Aging of Primary Human Embryonic Lung and Foreskin Fibroblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:731938. [PMID: 26339636 PMCID: PMC4538583 DOI: 10.1155/2015/731938] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 06/14/2015] [Accepted: 06/22/2015] [Indexed: 01/18/2023]
Abstract
Replicative senescence is of fundamental importance for the process of cellular aging, since it is a property of most of our somatic cells. Here, we elucidated this process by comparing gene expression changes, measured by RNA-seq, in fibroblasts originating from two different tissues, embryonic lung (MRC-5) and foreskin (HFF), at five different time points during their transition into senescence. Although the expression patterns of both fibroblast cell lines can be clearly distinguished, the similar differential expression of an ensemble of genes was found to correlate well with their transition into senescence, with only a minority of genes being cell line specific. Clustering-based approaches further revealed common signatures between the cell lines. Investigation of the mRNA expression levels at various time points during the lifespan of either of the fibroblasts resulted in a number of monotonically up- and downregulated genes which clearly showed a novel strong link to aging and senescence related processes which might be functional. In terms of expression profiles of differentially expressed genes with age, common genes identified here have the potential to rule the transition into senescence of embryonic lung and foreskin fibroblasts irrespective of their different cellular origin.
Collapse
|
37
|
Larkin J, Lohr TA, Elefante L, Shearin J, Matico R, Su JL, Xue Y, Liu F, Genell C, Miller RE, Tran PB, Malfait AM, Maier CC, Matheny CJ. Translational development of an ADAMTS-5 antibody for osteoarthritis disease modification. Osteoarthritis Cartilage 2015; 23:1254-66. [PMID: 25800415 PMCID: PMC4516626 DOI: 10.1016/j.joca.2015.02.778] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 02/03/2015] [Accepted: 02/24/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE/METHOD Aggrecanase activity, most notably ADAMTS-5, is implicated in pathogenic cartilage degradation. Selective monoclonal antibodies (mAbs) to both ADAMTS-5 and ADAMTS-4 were generated and in vitro, ex vivo and in vivo systems were utilized to assess target engagement, aggrecanase inhibition and modulation of disease-related endpoints with the intent of selecting a candidate for clinical development in osteoarthritis (OA). RESULTS Structural mapping predicts the most potent mAbs employ a unique mode of inhibition by cross-linking the catalytic and disintegrin domains. In a surgical mouse model of OA, both ADAMTS-5 and ADAMTS-4-specific mAbs penetrate cartilage following systemic administration, demonstrating access to the anticipated site of action. Structural disease modification and associated alleviation of pain-related behavior were observed with ADAMTS-5 mAb treatment. Treatment of human OA cartilage demonstrated a preferential role for ADAMTS-5 inhibition over ADAMTS-4, as measured by ARGS neoepitope release in explant cultures. ADAMTS-5 mAb activity was most evident in a subset of patient-derived tissues and suppression of ARGS neoepitope release was sustained for weeks after a single treatment in human explants and in cynomolgus monkeys, consistent with high affinity target engagement and slow ADAMTS-5 turnover. CONCLUSION This data supports a hypothesis set forth from knockout mouse studies that ADAMTS-5 is the major aggrecanase involved in cartilage degradation and provides a link between a biological pathway and pharmacology which translates to human tissues, non-human primate models and points to a target OA patient population. Therefore, a humanized ADAMTS-5-selective monoclonal antibody (GSK2394002) was progressed as a potential OA disease modifying therapeutic.
Collapse
Affiliation(s)
- Jonathan Larkin
- Experimental Medicine Unit – Immuno-Inflammation Therapeutic Area Unit, GlaxoSmithKline, Upper Merion, Pennsylvania, USA,Novel Targets Biopharm Discovery Unit – Biopharm R&D, GlaxoSmithKline, Upper Merion, Pennsylvania, USA,Corresponding author:
| | - Thomas A. Lohr
- Experimental Medicine Unit – Immuno-Inflammation Therapeutic Area Unit, GlaxoSmithKline, Upper Merion, Pennsylvania, USA,Novel Targets Biopharm Discovery Unit – Biopharm R&D, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Louis Elefante
- Novel Targets Biopharm Discovery Unit – Biopharm R&D, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Jean Shearin
- Biological Sciences, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Rosalie Matico
- Biological Sciences, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Jui-Lan Su
- Biological Sciences, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Yu Xue
- Novel Targets Biopharm Discovery Unit – Biopharm R&D, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Feng Liu
- Quantitative Sciences, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Caroline Genell
- ImmunoToxicology – Platform Technology & Science, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | - Rachel E. Miller
- Department of Internal Medicine (Rheumatology), Rush University Medical Center; Chicago, Illinois
| | - Phuong B. Tran
- Department of Internal Medicine (Rheumatology), Rush University Medical Center; Chicago, Illinois
| | - Anne-Marie Malfait
- Department of Internal Medicine (Rheumatology), Rush University Medical Center; Chicago, Illinois
| | - Curtis C. Maier
- ImmunoToxicology – Platform Technology & Science, GlaxoSmithKline, Upper Merion, Pennsylvania, USA
| | | |
Collapse
|
38
|
Dubail J, Apte SS. Insights on ADAMTS proteases and ADAMTS-like proteins from mammalian genetics. Matrix Biol 2015; 44-46:24-37. [PMID: 25770910 DOI: 10.1016/j.matbio.2015.03.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 01/05/2023]
Abstract
The mammalian ADAMTS superfamily comprises 19 secreted metalloproteinases and 7 ADAMTS-like proteins, each the product of a distinct gene. Thus far, all appear to be relevant to extracellular matrix function or to cell-matrix interactions. Most ADAMTS functions first emerged from analysis of spontaneous human and animal mutations and genetically engineered animals. The clinical manifestations of Mendelian disorders resulting from mutations in ADAMTS2, ADAMTS10, ADAMTS13, ADAMTS17, ADAMTSL2 and ADAMTSL4 identified essential roles for each gene, but also suggested potential cooperative functions of ADAMTS proteins. These observations were extended by analysis of spontaneous animal mutations, such as in bovine ADAMTS2, canine ADAMTS10, ADAMTS17 and ADAMTSL2 and mouse ADAMTS20. These human and animal disorders are recessive and their manifestations appear to result from a loss-of-function mechanism. Genome-wide analyses have determined an association of some ADAMTS loci such as ADAMTS9 and ADAMTS7, with specific traits and acquired disorders. Analysis of genetically engineered rodent mutations, now achieved for over half the superfamily, has provided novel biological insights and animal models for the respective human genetic disorders and suggested potential candidate genes for related human phenotypes. Engineered mouse mutants have been interbred to generate combinatorial mutants, uncovering cooperative functions of ADAMTS proteins in morphogenesis. Specific genetic models have provided crucial insights on mechanisms of osteoarthritis (OA), a common adult-onset degenerative condition. Engineered mutants will facilitate interpretation of exome variants identified in isolated birth defects and rare genetic conditions, as well as in genome-wide screens for trait and disease associations. Mammalian forward and reverse genetics, together with genome-wide analysis, together constitute a powerful force for revealing the functions of ADAMTS proteins in physiological pathways and health disorders. Their continuing use, together with genome-editing technology and the ability to generate stem cells from mutants, presents numerous opportunities for advancing basic knowledge, human disease pathways and therapy.
Collapse
Affiliation(s)
- Johanne Dubail
- Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Suneel S Apte
- Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
39
|
Smith MM, Melrose J. Proteoglycans in Normal and Healing Skin. Adv Wound Care (New Rochelle) 2015; 4:152-173. [PMID: 25785238 DOI: 10.1089/wound.2013.0464] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Indexed: 02/04/2023] Open
Abstract
Significance: Proteoglycans have a distinct spatial localization in normal skin and are essential for the correct structural development, organization, hydration, and functional properties of this tissue. The extracellular matrix (ECM) is no longer considered to be just an inert supportive material but is a source of directive, spatial and temporal, contextual information to the cells via components such as the proteoglycans. There is a pressing need to improve our understanding of how these important molecules functionally interact with other matrix structures, cells and cellular mediators in normal skin and during wound healing. Recent Advances: New antibodies to glycosaminoglycan side chain components of skin proteoglycans have facilitated the elucidation of detailed localization patterns within skin. Other studies have revealed important proliferative activities of proteinase-generated fragments of proteoglycans and other ECM components (matricryptins). Knockout mice have further established the functional importance of skin proteoglycans in the assembly and homeostasis of the normal skin ECM. Critical Issues: Our comprehension of the molecular and structural complexity of skin as a complex, dynamic, constantly renewing, layered connective tissue is incomplete. The impact of changes in proteoglycans on skin pathology and the wound healing process is recognized as an important area of pathobiology and is an area of intense investigation. Future Directions: Advanced technology is allowing the development of new artificial skins. Recent knowledge on skin proteoglycans can be used to incorporate these molecules into useful adjunct therapies for wound healing and for maintenance of optimal tissue homeostasis in aging skin.
Collapse
Affiliation(s)
- Margaret Mary Smith
- Raymond Purves Research Laboratories, Kolling Institute (University of Sydney), Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - James Melrose
- Raymond Purves Research Laboratories, Kolling Institute (University of Sydney), Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
40
|
Cipriani P, Ruscitti P, Di Benedetto P, Carubbi F, Liakouli V, Berardicurti O, Ciccia F, Triolo G, Giacomelli R. Mesenchymal stromal cells and rheumatic diseases: new tools from pathogenesis to regenerative therapies. Cytotherapy 2015; 17:832-49. [PMID: 25680301 DOI: 10.1016/j.jcyt.2014.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/24/2014] [Accepted: 12/01/2014] [Indexed: 01/08/2023]
Abstract
In recent years, mesenchymal stromal cells (MSCs) have been largely investigated and tested as a new therapeutic tool for several clinical applications, including the treatment of different rheumatic diseases. MSCs are responsible for the normal turnover and maintenance of adult mesenchymal tissues as the result of their multipotent differentiation abilities and their secretion of a variety of cytokines and growth factors. Although initially derived from bone marrow, MSCs are present in many different tissues such as many peri-articular tissues. MSCs may exert immune-modulatory properties, modulating different immune cells in both in vitro and in vivo models, and they are considered immune-privileged cells. At present, these capacities are considered the most intriguing aspect of their biology, introducing the possibility that these cells may be used as effective therapy in autoimmune diseases. Therefore, stem cell therapies may represent an innovative approach for the treatment of rheumatic diseases, especially for the forms that are not responsive to standard treatments or alternatively still lacking a definite therapy. At present, although the data from scientific literature appear to suggest that such treatments might be more effective whether administered as soon as possible, the use of MSCs in clinical practice is likely to be restricted to patients with a long history of a severe refractory disease. Further results from larger clinical trials are needed to corroborate preclinical findings and human non-controlled studies, and advancement in the knowledge of MSCs might provide information about the therapeutic role of these cells in the treatment of many rheumatic diseases.
Collapse
Affiliation(s)
- Paola Cipriani
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy.
| | - Piero Ruscitti
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Paola Di Benedetto
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Francesco Carubbi
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Vasiliki Liakouli
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Onorina Berardicurti
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Francesco Ciccia
- Rheumatology Unit, Internal Medicine Department, University of Palermo, Palermo, Italy
| | - Giovanni Triolo
- Rheumatology Unit, Internal Medicine Department, University of Palermo, Palermo, Italy
| | - Roberto Giacomelli
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
41
|
Bell R, Li J, Shewman EF, Galante JO, Cole BJ, Bach BR, Troy KL, Mikecz K, Sandy JD, Plaas AH, Wang VM. ADAMTS5 is required for biomechanically-stimulated healing of murine tendinopathy. J Orthop Res 2013; 31:1540-8. [PMID: 23754494 DOI: 10.1002/jor.22398] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 05/01/2013] [Indexed: 02/04/2023]
Abstract
A recently developed murine model of tendinopathy, induced by TGF-β1 injection, has been used to examine the reparative capacity of tendinopathic Achilles in Adamts5(-/-) mice. After TGF-β1 injection and 2 weeks of treadmill exercise, the Achilles from Adamts5(-/-) mice exhibited a reduction in maximum tensile stress of approximately 60%. However, in contrast to wild type mice previously characterized by this model, Adamts5(-/-) mice subjected to further treadmill exercise were unable to reverse this biomechanical deficit. This nonreparative phenotype was accompanied by a major deficiency, relative to wild-type, in expression of Col1a1 and Col3a1 and an abnormally elevated expression of a wide range of integrins. In addition, the tendinopathic Adamts5(-/-) mice showed a persistent accumulation of chondrogenic cells in the tendon body and an aggrecan-rich fibrocartilaginous matrix within disorganized collagen fiber bundles. Moreover, consistent with the compromised biomechanical properties of the Achilles in the Adamts5(-/-) mice, in vivo gait analysis revealed a strong trend (p = 0.07) towards increased swing time of the injected limb in Adamts5(-/-) relative to wild-type mice. These findings demonstrate that a deficiency in ADAMTS5 promotes a chondrogenic response to TGF-β1 injection that is not reversed by treadmill exercise. Hence, repair of biomechanically compromised tendons exhibiting midsubstance chondroid accumulation requires ADAMTS5.
Collapse
Affiliation(s)
- Rebecca Bell
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W. Harrison Street, Suite 201, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Osteoarthritis (OA), a prevalent chronic condition with a striking impact on quality of life, represents an enormous societal burden that increases greatly as populations age. Yet no approved pharmacological intervention, biologic therapy or procedure prevents the progressive destruction of the OA joint. Mesenchymal stem cells (MSCs)-multipotent precursors of connective tissue cells that can be isolated from many adult tissues, including those of the diarthrodial joint-have emerged as a potential therapy. Endogenous MSCs contribute to maintenance of healthy tissues by acting as reservoirs of repair cells or as immunomodulatory sentinels to reduce inflammation. The onset of degenerative changes in the joint is associated with aberrant activity or depletion of these cell reservoirs, leading to loss of chondrogenic potential and preponderance of a fibrogenic phenotype. Local delivery of ex vivo cultures of MSCs has produced promising outcomes in preclinical models of joint disease. Mechanistically, paracrine signalling by MSCs might be more important than differentiation in stimulating repair responses; thus, paracrine factors must be assessed as measures of MSC therapeutic potency, to replace traditional assays based on cell-surface markers and differentiation. Several early-stage clinical trials, initiated or underway in 2013, are testing the delivery of MSCs as an intra-articular injection into the knee, but optimal dose and vehicle are yet to be established.
Collapse
|
43
|
Abstract
Transforming Growth Factor-β (TGF-β) superfamily ligands regulate many aspects of cell identity, function, and survival in multicellular animals. Genes encoding five TGF-β family members are present in the genome of C. elegans. Two of the ligands, DBL-1 and DAF-7, signal through a canonical receptor-Smad signaling pathway; while a third ligand, UNC-129, interacts with a noncanonical signaling pathway. No function has yet been associated with the remaining two ligands. Here we summarize these signaling pathways and their biological functions.
Collapse
Affiliation(s)
- Tina L Gumienny
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX 77843, USA
| | | |
Collapse
|
44
|
Jones ER, Jones GC, Legerlotz K, Riley GP. Cyclical strain modulates metalloprotease and matrix gene expression in human tenocytes via activation of TGFβ. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2596-2607. [PMID: 23830915 PMCID: PMC3898605 DOI: 10.1016/j.bbamcr.2013.06.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 06/17/2013] [Accepted: 06/20/2013] [Indexed: 12/11/2022]
Abstract
Tendinopathies are a range of diseases characterised by degeneration and chronic tendon pain and represent a significant cause of morbidity. Relatively little is known about the underlying mechanisms; however onset is often associated with physical activity. A number of molecular changes have been documented in tendinopathy such as a decrease in overall collagen content, increased extracellular matrix turnover and protease activity. Metalloproteinases are involved in the homeostasis of the extracellular matrix and expression is regulated by mechanical strain. The aims of this study were to determine the effects of strain upon matrix turnover by measuring metalloproteinase and matrix gene expression and to elucidate the mechanism of action. Primary Human Achilles tenocytes were seeded in type I rat tail collagen gels in a Flexcell™ tissue train system and subjected to 5% cyclic uniaxial strain at 1 Hz for 48 h. TGFβ1 and TGFβRI inhibitor were added to selected cultures. RNA was measured using qRT-PCR and TGFβ protein levels were determined using a cell based luciferase assay. We observed that mechanical strain regulated the mRNA levels of multiple protease and matrix genes anabolically, and this regulation mirrored that seen with TGFβ stimulation alone. We have also demonstrated that the inhibition of the TGFβ signalling pathway abrogated the strain induced changes in mRNA and that TGFβ activation, rather than gene expression, was increased with mechanical strain. We concluded that TGFβ activation plays an important role in mechanotransduction. Targeting this pathway may have its place in the treatment of tendinopathy. Mechanical strain regulates multiple protease and matrix genes at the mRNA level. Changes in mRNA level are analogous to those induced by TGFβ stimulation. The inhibition of the TGFβ signalling pathway abrogated the strain-induced changes. A SMAD activatory soluble factor is increased in activity in response to mechanical load.
Collapse
Affiliation(s)
- Eleanor R Jones
- Soft Tissue Research Group, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK.
| | - Gavin C Jones
- Soft Tissue Research Group, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Kirsten Legerlotz
- Soft Tissue Research Group, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Graham P Riley
- Soft Tissue Research Group, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| |
Collapse
|
45
|
Yi JH, Katagiri Y, Susarla B, Figge D, Symes AJ, Geller HM. Alterations in sulfated chondroitin glycosaminoglycans following controlled cortical impact injury in mice. J Comp Neurol 2013; 520:3295-313. [PMID: 22628090 DOI: 10.1002/cne.23156] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) play a pivotal role in many neuronal growth mechanisms including axon guidance and the modulation of repair processes following injury to the spinal cord or brain. Many actions of CSPGs in the central nervous system (CNS) are governed by the specific sulfation pattern on the glycosaminoglycan (GAG) chains attached to CSPG core proteins. To elucidate the role of CSPGs and sulfated GAG chains following traumatic brain injury (TBI), controlled cortical impact injury of mild to moderate severity was performed over the left sensory motor cortex in mice. Using immunoblotting and immunostaining, we found that TBI resulted in an increase in the CSPGs neurocan and NG2 expression in a tight band surrounding the injury core, which overlapped with the presence of 4-sulfated CS GAGs but not with 6-sulfated GAGs. This increase was observed as early as 7 days post injury (dpi), and persisted for up to 28 dpi. Labeling with markers against microglia/macrophages, NG2+ cells, fibroblasts, and astrocytes showed that these cells were all localized in the area, suggesting multiple origins of chondroitin-4-sulfate increase. TBI also caused a decrease in the expression of aggrecan and phosphacan in the pericontusional cortex with a concomitant reduction in the number of perineuronal nets. In summary, we describe a dual response in CSPGs whereby they may be actively involved in complex repair processes following TBI.
Collapse
Affiliation(s)
- Jae-Hyuk Yi
- Developmental Neurobiology Section, Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
46
|
Insufficient versican cleavage and Smad2 phosphorylation results in bicuspid aortic and pulmonary valves. J Mol Cell Cardiol 2013; 60:50-9. [PMID: 23531444 DOI: 10.1016/j.yjmcc.2013.03.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 03/10/2013] [Accepted: 03/11/2013] [Indexed: 12/29/2022]
Abstract
Bicuspid or bifoliate aortic valve (BAV) results in two rather than three cusps and occurs in 1-2% of the population placing them at higher risk of developing progressive aortic valve disease. Only NOTCH-1 has been linked to human BAV, and genetically modified mouse models of BAV are limited by low penetrance and additional malformations. Here we report that in the Adamts5(-/-) valves, collagen I, collagen III, and elastin were disrupted in the malformed hinge region that anchors the mature semilunar cusps and where the ADAMTS5 proteoglycan substrate versican, accumulates. ADAMTS5 deficient prevalvular mesenchyme also exhibited a reduction of α-smooth muscle actin and filamin A suggesting versican cleavage may be involved in TGFβ signaling. Subsequent evaluation showed a significant decrease of pSmad2 in regions of prevalvular mesenchyme in Adamts5(-/-) valves. To test the hypothesis that ADAMTS5 versican cleavage is required, in part, to elicit Smad2 phosphorylation we further reduced Smad2 in Adamts5(-/-) mice through intergenetic cross. The Adamts5(-/-);Smad2(+/-) mice had highly penetrant BAV and bicuspid pulmonary valve (BPV) malformations as well as increased cusp and hinge size compared to the Adamts5(-/-) and control littermates. These studies demonstrate that semilunar cusp malformations (BAV and BPV) can arise from a failure to remodel the proteoglycan-rich provisional ECM. Specifically, faulty versican clearance due to ADAMTS5 deficiency blocks the initiation of pSmad2 signaling, which is required for excavation of endocardial cushions during aortic and pulmonary valve development. Further studies using the Adamts5(-/-); Smad2(+/-) mice with highly penetrant and isolated BAV, may lead to new pharmacological treatments for valve disease.
Collapse
|
47
|
Pulsatelli L, Addimanda O, Brusi V, Pavloska B, Meliconi R. New findings in osteoarthritis pathogenesis: therapeutic implications. Ther Adv Chronic Dis 2013; 4:23-43. [PMID: 23342245 DOI: 10.1177/2040622312462734] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This review focuses on the new perspectives which can provide insight into the crucial pathways that drive cartilage-bone physiopathology. In particular, we discuss the critical signaling and effector molecules that can activate cellular and molecular processes in both cartilage and bone cells and which may be relevant in cross talk among joint compartments: growth factors (bone morphogenetic proteins and transforming growth factor), hypoxia-related factors, cell-matrix interactions [discoidin domain receptor 2 (DDR2) and syndecan 4], signaling molecules [WNT, Hedgehog (Hh)]. With the continuous progression of our knowledge on the molecular pathways involved in cartilage and bone changes in osteoarthritis (OA), an increasing number of potentially effective candidates for OA therapy are already under scrutiny in clinical trials to ascertain their possible safe use in an attempt to identify molecules active in slowing or halting OA progression and reducing joint pain. We then review the principal molecules currently under clinical investigation.
Collapse
Affiliation(s)
- Lia Pulsatelli
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, Bologna, Italy
| | | | | | | | | |
Collapse
|
48
|
Stupka N, Kintakas C, White JD, Fraser FW, Hanciu M, Aramaki-Hattori N, Martin S, Coles C, Collier F, Ward AC, Apte SS, McCulloch DR. Versican processing by a disintegrin-like and metalloproteinase domain with thrombospondin-1 repeats proteinases-5 and -15 facilitates myoblast fusion. J Biol Chem 2012; 288:1907-17. [PMID: 23233679 DOI: 10.1074/jbc.m112.429647] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Skeletal muscle development and regeneration requires the fusion of myoblasts into multinucleated myotubes. Because the enzymatic proteolysis of a hyaluronan and versican-rich matrix by ADAMTS versicanases is required for developmental morphogenesis, we hypothesized that the clearance of versican may facilitate the fusion of myoblasts during myogenesis. Here, we used transgenic mice and an in vitro model of myoblast fusion, C2C12 cells, to determine a potential role for ADAMTS versicanases. Versican processing was observed during in vivo myogenesis at the time when myoblasts were fusing to form multinucleated myotubes. Relevant ADAMTS genes, chief among them Adamts5 and Adamts15, were expressed both in developing embryonic muscle and differentiating C2C12 cells. Reducing the levels of Adamts5 mRNA in vitro impaired myoblast fusion, which could be rescued with catalytically active but not the inactive forms of ADAMTS5 or ADAMTS15. The addition of inactive ADAMTS5, ADAMTS15, or full-length V1 versican effectively impaired myoblast fusion. Finally, the expansion of a hyaluronan and versican-rich matrix was observed upon reducing the levels of Adamts5 mRNA in myoblasts. These data indicate that these ADAMTS proteinases contribute to the formation of multinucleated myotubes such as is necessary for both skeletal muscle development and during regeneration, by remodeling a versican-rich pericellular matrix of myoblasts. Our study identifies a possible pathway to target for the improvement of myogenesis in a plethora of diseases including cancer cachexia, sarcopenia, and muscular dystrophy.
Collapse
Affiliation(s)
- Nicole Stupka
- School of Medicine and Molecular and Medical Research SRC, Deakin University, Geelong, Victoria 3216, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kumar S, Rao N, Ge R. Emerging Roles of ADAMTSs in Angiogenesis and Cancer. Cancers (Basel) 2012; 4:1252-99. [PMID: 24213506 PMCID: PMC3712723 DOI: 10.3390/cancers4041252] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/21/2012] [Accepted: 11/23/2012] [Indexed: 12/18/2022] Open
Abstract
A Disintegrin-like And Metalloproteinase with ThromboSpondin motifs—ADAMTSs—are a multi-domain, secreted, extracellular zinc metalloproteinase family with 19 members in humans. These extracellular metalloproteinases are known to cleave a wide range of substrates in the extracellular matrix. They have been implicated in various physiological processes, such as extracellular matrix turnover, melanoblast development, interdigital web regression, blood coagulation, ovulation, etc. ADAMTSs are also critical in pathological processes such as arthritis, atherosclerosis, cancer, angiogenesis, wound healing, etc. In the past few years, there has been an explosion of reports concerning the role of ADAMTS family members in angiogenesis and cancer. To date, 10 out of the 19 members have been demonstrated to be involved in regulating angiogenesis and/or cancer. The mechanism involved in their regulation of angiogenesis or cancer differs among different members. Both angiogenesis-dependent and -independent regulation of cancer have been reported. This review summarizes our current understanding on the roles of ADAMTS in angiogenesis and cancer and highlights their implications in cancer therapeutic development.
Collapse
Affiliation(s)
- Saran Kumar
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| | | | | |
Collapse
|
50
|
Bell R, Li J, Gorski DJ, Bartels AK, Shewman EF, Wysocki RW, Cole BJ, Bach BR, Mikecz K, Sandy JD, Plaas AH, Wang VM. Controlled treadmill exercise eliminates chondroid deposits and restores tensile properties in a new murine tendinopathy model. J Biomech 2012; 46:498-505. [PMID: 23159096 DOI: 10.1016/j.jbiomech.2012.10.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/08/2012] [Accepted: 10/21/2012] [Indexed: 11/25/2022]
Abstract
Tendinopathy is a widespread and disabling condition characterized by collagen fiber disruption and accumulation of a glycosaminoglycan-rich chondroid matrix. Recent clinical reports have illustrated the potential of mechanical loading (exercise) therapies to successfully treat chronic tendinopathies. We have developed a new murine tendinopathy model which requires a single injection of TGF-β1 into the Achilles tendon midsubstance followed by normal cage activity for 2 weeks. At this time, tendon maximum stress showed a dramatic (66%) reduction relative to that of normal controls and this persisted at four weeks. Loss of material properties was accompanied by abundant chondroid cells within the tendon (closely resembling the changes observed in human samples obtained intra-operatively) and increased expression of Acan, Col1a1, Col2a1, Col3a1, Fn1 and Mmp3. Mice subjected to two weeks of daily treadmill exercise following TGF-β1 injection showed a similar reduction in tendon material properties as the caged group. However, in mice subjected to 4 weeks of treadmill exercise, tendon maximum stress values were similar to those of naive controls. Tendons from the mice exercised for 4 weeks showed essentially no chondroid cells and the expression of Acan, Col1a1, Col2a1, Col3a1, and Mmp3 was significantly reduced relative to the 4-week cage group. This technically simple murine tendinopathy model is highly amenable to detailed mechanistic and translational studies of the biomechanical and cell biological pathways, that could be targeted to enhance healing of tendinopathy.
Collapse
Affiliation(s)
- Rebecca Bell
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|